1
|
Lampros M, Alexiou GA. Brain and Spinal Cord Tumors of Embryonic Origin. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1405:405-420. [PMID: 37452947 DOI: 10.1007/978-3-031-23705-8_15] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/18/2023]
Abstract
Embryonal tumors (ETs) of the central nervous system (CNS) comprise a large heterogeneous group of highly malignant tumors that predominantly affect children and adolescents. Currently, the neoplasms classified as ET are the medulloblastoma (MB), embryonal tumors with multilayered rosettes (ETMR), medulloepithelioma (ME), CNS neuroblastoma (NB), CNS ganglioneuroblastoma (GNB), atypical teratoid/rhabdoid tumors (AT/RT), and CNS embryonal tumors with rhabdoid features. All these tumors are classified as malignant-grade IV neoplasms, and the prognosis of patients with these neoplasms is very poor. Currently, except for the histological classification of MB, the recently utilized WHO classification accepts a novel molecular classification of MBs into four distinct molecular subgroups: wingless/integrated (WNT)-activated, sonic hedgehog (Shh), and the numerical Group3 and Group 4. The combination of both histological and genetic classifications has substantial prognostic significance, and patients are categorized as low risk with over 90% survival, the standard risk with 75-90% survival, high risk with 50-75% survival, and very high risk with survival rate lower than 50%. Children under three years are predominantly affected by AT/RT and represent about 20% of all CNS tumors in this age group. AT/RT is typically located in the posterior fossa (mainly in cerebellopontine angle) in 50-60% of the cases. The pathogenesis of this neoplasm is strongly associated with loss of function of the SMARCB1 (INI1, hSNF5) gene located at the 22q11.23 chromosome, or very rarely with alterations in (SMARCA4) BRG1 gene. The cells of this neoplasm resemble those of other neuronal tumors, and hence, immunochemistry markers have been utilized, such as smooth muscle actin, epithelial membrane antigen, vimentin, and lately antibodies for INI1. ETMRs are characterized by the presence of ependymoblastic rosettes formed by undifferentiated neuroepithelial cells and neuropil. The tumorigenesis of ETMRs is strongly related to the amplification of the pluripotency factor Chr19q13.41 miRNA cluster (C19MC) present in around 90% of the cases. Additionally, the expression of LIN28A is a highly sensitive and specific marker of ETMR diagnosis, as it is overexpressed in almost all cases of ETMR and is related to poor patient outcomes. The treatment of patients with ETs includes a combination of surgical resection, radiotherapy (focal or craniospinal), and chemotherapeutic agents. Currently, there is a trend to reduce the dose of craniospinal irradiation in the treatment of low-risk MBs. Novel targeted therapies are expected in the treatment of patients with MBs due to the identification of the main driver genes. Survival rates vary between ET types and their subtypes, with ganglioneuroblastoma having over 95% 5-year survival rate, while ATRT is probably linked with the worst prognosis with a 30% 5-year survival rate.
Collapse
Affiliation(s)
- Marios Lampros
- Department of Neurosurgery, University Hospital of Ioannina, Ioannina, Greece
| | - George A Alexiou
- Department of Neurosurgery, School of Medicine, University of Ioannina, 45500, Ioannina, Greece.
| |
Collapse
|
2
|
Whitney J, Dollinger L, Tamrazi B, Hawes D, Couce M, Marcheque J, Judkins A, Margol A, Madabhushi A. Quantitative Nuclear Histomorphometry Predicts Molecular Subtype and Clinical Outcome in Medulloblastomas: Preliminary Findings. J Pathol Inform 2022; 13:100090. [PMID: 36268104 PMCID: PMC9576985 DOI: 10.1016/j.jpi.2022.100090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Accepted: 12/11/2021] [Indexed: 10/30/2022] Open
Abstract
Molecular subtypes of medulloblastoma [Sonic Hedgehog (SHH), Wingless/INT (WNT), Group 3, and Group 4] are defined by common patterns of gene expression. These differential gene expression patterns appear to result in different histomorphology and prognosis. Quantitative histomorphometry is a well-known method of computer-aided pathology image analysis. The hypotheses we sought to examine in this preliminary proof of concept study were whether computer extracted nuclear morphological features of medulloblastomas from digitized tissue slide images could independently: (1) distinguish between molecularly determined subgroups and (2) identify patterns within these subgroups that correspond with clinical outcome. Our dataset was composed of 46 medulloblastoma patients: 16 SHH (5 dead, 11 survived), 3 WNT (0 dead, 3 survived), 12 Group 3 (4 dead, 8 survived), and 15 were Group 4 (5 dead, 10 survived). A watershed-based thresholding scheme was used to automatically identify individual nuclei within digitized whole slide hematoxylin and eosin tissue images. Quantitative histomorphometric features corresponding to the texture (variation in pixel intensity), shape (variations in size, roundness), and architectural rearrangement (distances between, and number of connected neighbors) of nuclei were subsequently extracted. These features were ranked using feature selection schemes and these top-ranked features were then used to train machine-learning classifiers via threefold cross-validation to separate patients based on: (1) molecular subtype and (2) disease-specific outcomes within the individual molecular subtype groups. SHH and WNT tumors were separated from Groups 3 and 4 tumors with a maximum area under the receiver operating characteristic curve (AUC) of 0.7, survival within Group 3 tumors was predicted with an AUC of 0.92, and Group 3 and 4 patients were separated into high- and low-risk groups with p = 0.002. Model prediction was quantitatively compared with age, stage, and histological subtype using univariate and multivariate Cox hazard ratio models. Age was the most statistically significant variable for predicting survival in Group 3 and 4 tumors, but model predictions had the highest hazard ratio value. Quantitative nuclear histomorphometry can be used to study medulloblastoma genetic expression phenotypes as it may distinguish meaningful features of disease pathology.
Collapse
|
3
|
Xiong A, Spyrou A, Forsberg-Nilsson K. Involvement of Heparan Sulfate and Heparanase in Neural Development and Pathogenesis of Brain Tumors. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1221:365-403. [PMID: 32274718 DOI: 10.1007/978-3-030-34521-1_14] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Brain tumors are aggressive and devastating diseases. The most common type of brain tumor, glioblastoma (GBM), is incurable and has one of the worst five-year survival rates of all human cancers. GBMs are invasive and infiltrate healthy brain tissue, which is one main reason they remain fatal despite resection, since cells that have already migrated away lead to rapid regrowth of the tumor. Curative therapy for medulloblastoma (MB), the most common pediatric brain tumor, has improved, but the outcome is still poor for many patients, and treatment causes long-term complications. Recent advances in the classification of pediatric brain tumors reveal distinct subgroups, allowing more targeted therapy for the most aggressive forms, and sparing children with less malignant tumors the side-effects of massive treatment. Heparan sulfate proteoglycans (HSPGs), main components of the neurogenic niche, interact specifically with a large number of physiologically important molecules and vital roles for HS biosynthesis and degradation in neural stem cell differentiation have been presented. HSPGs are composed of a core protein with attached highly charged, sulfated disaccharide chains. The major enzyme that degrades HS is heparanase (HPSE), an important regulator of extracellular matrix (ECM) remodeling which has been suggested to promote the growth and invasion of other types of tumors. This is of clinical interest because GBM are highly invasive and children with metastatic MB at the time of diagnosis exhibit a worse outcome. Here we review the involvement of HS and HPSE in development of the nervous system and some of its most malignant brain tumors, glioblastoma and medulloblastoma.
Collapse
Affiliation(s)
- Anqi Xiong
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
- Department of Medical Biochemistry and Biophysics, Karolinska Insitutet, Stockholm, Sweden
| | - Argyris Spyrou
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Karin Forsberg-Nilsson
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, Uppsala, Sweden.
| |
Collapse
|
4
|
Hanaford AR, Alt J, Rais R, Wang SZ, Kaur H, Thorek DLJ, Eberhart CG, Slusher BS, Martin AM, Raabe EH. Orally bioavailable glutamine antagonist prodrug JHU-083 penetrates mouse brain and suppresses the growth of MYC-driven medulloblastoma. Transl Oncol 2019; 12:1314-1322. [PMID: 31340195 PMCID: PMC6657308 DOI: 10.1016/j.tranon.2019.05.013] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Revised: 05/13/2019] [Accepted: 05/13/2019] [Indexed: 12/22/2022] Open
Abstract
A subset of poor-prognosis medulloblastoma has genomic amplification of MYC. MYC regulates glutamine metabolism in multiple cellular contexts. We modified the glutamine analog 6-diazo-5-oxo-l-norleucine (DON) to mask its carboxylate and amine functionalities, creating a prodrug termed JHU-083 with increased oral bioavailability. We hypothesized that this prodrug would kill MYC-expressing medulloblastoma. JHU-083 treatment caused decreased growth and increased apoptosis in human MYC-expressing medulloblastoma cell lines. We generated a mouse MYC-driven medulloblastoma model by transforming C57BL/6 mouse cerebellar stem and progenitor cells. When implanted into the brains of C57BL/6 mice, these cells formed large cell/anaplastic tumors that resembled aggressive medulloblastoma. A cell line derived from this model was sensitive to JHU-083 in vitro. Oral administration of JHU-038 led to the accumulation of micromolar concentrations of DON in the mouse brain. JHU-083 treatment significantly increased the survival of immune-competent animals bearing orthotopic tumors formed by the mouse cerebellar stem cell model as well as immune-deficient animals bearing orthotopic tumors formed by a human MYC-amplified medulloblastoma cell line. These data provide pre-clinical justification for the ongoing development and testing of orally bioavailable DON prodrugs for use in medulloblastoma patients.
Collapse
Affiliation(s)
- Allison R Hanaford
- Division of Pediatric Oncology, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Jesse Alt
- Johns Hopkins Drug Discovery, Johns Hopkins University School of Medicine
| | - Rana Rais
- Johns Hopkins Drug Discovery, Johns Hopkins University School of Medicine; Department of Neurology, Johns Hopkins University School of Medicine
| | - Sabrina Z Wang
- Division of Pediatric Oncology, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Harpreet Kaur
- Division of Pediatric Oncology, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Daniel L J Thorek
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO; Department of Biomedical Engineering, Washington University, St. Louis, MO
| | - Charles G Eberhart
- Department of Pathology, Johns Hopkins University, School of Medicine, Baltimore, MD 21287; Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, School of Medicine, Baltimore, MD 21287
| | - Barbara S Slusher
- Johns Hopkins Drug Discovery, Johns Hopkins University School of Medicine; Department of Neurology, Johns Hopkins University School of Medicine
| | - Allison M Martin
- Division of Pediatric Oncology, Johns Hopkins University School of Medicine, Baltimore, MD; Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, School of Medicine, Baltimore, MD 21287.
| | - Eric H Raabe
- Division of Pediatric Oncology, Johns Hopkins University School of Medicine, Baltimore, MD; Department of Pathology, Johns Hopkins University, School of Medicine, Baltimore, MD 21287; Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, School of Medicine, Baltimore, MD 21287.
| |
Collapse
|
5
|
Abstract
Medulloblastoma (MB) comprises a biologically heterogeneous group of embryonal tumours of the cerebellum. Four subgroups of MB have been described (WNT, sonic hedgehog (SHH), Group 3 and Group 4), each of which is associated with different genetic alterations, age at onset and prognosis. These subgroups have broadly been incorporated into the WHO classification of central nervous system tumours but still need to be accounted for to appropriately tailor disease risk to therapy intensity and to target therapy to disease biology. In this Primer, the epidemiology (including MB predisposition), molecular pathogenesis and integrative diagnosis taking histomorphology, molecular genetics and imaging into account are reviewed. In addition, management strategies, which encompass surgical resection of the tumour, cranio-spinal irradiation and chemotherapy, are discussed, together with the possibility of focusing more on disease biology and robust molecularly driven patient stratification in future clinical trials.
Collapse
|
6
|
Huang PI, Lin SC, Lee YY, Ho DMT, Guo WY, Chang KP, Chang FC, Liang ML, Chen HH, Liu YM, Yen SH, Wong TT, Chen YW. Large cell/anaplastic medulloblastoma is associated with poor prognosis-a retrospective analysis at a single institute. Childs Nerv Syst 2017; 33:1285-1294. [PMID: 28488086 DOI: 10.1007/s00381-017-3435-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Accepted: 04/26/2017] [Indexed: 10/19/2022]
Abstract
PURPOSE Medulloblastoma (MB) is the most commonly occurring malignant pediatric brain tumor worldwide. However, a recent study found that the treatment outcomes in those with high-risk disease receiving conventional treatment were suboptimal. This study aimed to assess outcomes and treatment strategies for specific histologic subtypes of pediatric MB. METHODS A total of 114 pediatric patients (age < 20 years) diagnosed with MB between March 1998 and August 2011 were retrospectively reviewed; 52 that were treated with surgery followed by adjuvant radiotherapy (RT) and chemotherapy (CHT) were included. RESULTS The 5-year overall survival (OS) and relapse-free survival (RFS) rates were 73 and 69%, respectively. Median time to relapse was 17 months with a median survival time of 6 months after relapse. Patients of average risk had a better 5-year OS rate compared with high-risk patients (p = 0.027). The 5-year RFS of high-risk patients was lower compared with average risk (p = 0.038). A greater proportion of patients with large cell/anaplastic (LC/A) MB had recurrence than classic MB with 5-year RFS rate of 34 and 76%, respectively (p = 0.001), and OS rate of 56 and 76%, respectively (p = 0.04). CONCLUSION High-risk group and histology of LC/A were the most significant factors associated with worse OS and RFS. Patients with LC/A-MB had higher relapse rates and worse survival than those with classic MB. LC/A-MB carries a high risk for recurrence and should be treated with the more aggressive strategies.
Collapse
Affiliation(s)
- Pin-I Huang
- Division of Radiotherapy, Department of Oncology, Taipei Veterans General Hospital, 201, Sec. 2, Shih-Pai Road, Taipei, 11217, Taiwan.,Institute of Clinical Medicine, National Yang-Ming University, Taipei, Taiwan.,Faculty of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Shih-Chieh Lin
- Faculty of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Yi-Yen Lee
- Faculty of Medicine, National Yang-Ming University, Taipei, Taiwan.,Division of Pediatric Neurosurgery, Neurological Institute, Taipei Veterans General Hospital, 201, Sec. 2, Shih-Pai Road, Taipei, 11217, Taiwan
| | - Donald Ming-Tak Ho
- Faculty of Medicine, National Yang-Ming University, Taipei, Taiwan.,Department of Pathology, Taipei Veterans General Hospital, 201, Sec. 2, Shih-Pai Road, Taipei, 11217, Taiwan
| | - Wan-Yuo Guo
- Faculty of Medicine, National Yang-Ming University, Taipei, Taiwan.,Department of Radiology, Taipei Veterans General Hospital, 201, Sec. 2, Shih-Pai Road, Taipei, 11217, Taiwan
| | - Kai-Ping Chang
- Faculty of Medicine, National Yang-Ming University, Taipei, Taiwan.,Department of Pediatrics, Taipei Veterans General Hospital, 201, Sec. 2, Shih-Pai Road, Taipei, 11217, Taiwan
| | - Feng-Chi Chang
- Faculty of Medicine, National Yang-Ming University, Taipei, Taiwan.,Department of Radiology, Taipei Veterans General Hospital, 201, Sec. 2, Shih-Pai Road, Taipei, 11217, Taiwan
| | - Muh-Lii Liang
- Faculty of Medicine, National Yang-Ming University, Taipei, Taiwan.,Division of Pediatric Neurosurgery, Neurological Institute, Taipei Veterans General Hospital, 201, Sec. 2, Shih-Pai Road, Taipei, 11217, Taiwan
| | - Hsin-Hung Chen
- Faculty of Medicine, National Yang-Ming University, Taipei, Taiwan.,Division of Pediatric Neurosurgery, Neurological Institute, Taipei Veterans General Hospital, 201, Sec. 2, Shih-Pai Road, Taipei, 11217, Taiwan
| | - Yu-Ming Liu
- Division of Radiotherapy, Department of Oncology, Taipei Veterans General Hospital, 201, Sec. 2, Shih-Pai Road, Taipei, 11217, Taiwan.,Faculty of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Sang-Hue Yen
- Division of Radiotherapy, Department of Oncology, Taipei Veterans General Hospital, 201, Sec. 2, Shih-Pai Road, Taipei, 11217, Taiwan.,Department of Biomedical Imaging and Radiological Sciences, National Yang-Ming University, Taipei, Taiwan
| | - Tai-Tong Wong
- Faculty of Medicine, National Yang-Ming University, Taipei, Taiwan.,Division of Pediatric Neurosurgery, Neurological Institute, Taipei Veterans General Hospital, 201, Sec. 2, Shih-Pai Road, Taipei, 11217, Taiwan
| | - Yi-Wei Chen
- Division of Radiotherapy, Department of Oncology, Taipei Veterans General Hospital, 201, Sec. 2, Shih-Pai Road, Taipei, 11217, Taiwan. .,Faculty of Medicine, National Yang-Ming University, Taipei, Taiwan.
| |
Collapse
|
7
|
Abstract
Devil facial tumor disease (DFTD) is an emergent transmissible cancer exclusive to Tasmanian devils (Sarcophilus harrisii) and threatening the species with extinction in the wild. Research on DFTD began 10 years ago, when nothing was known about the tumor and little about the devils. The depth of knowledge gained since then is impressive, with research having addressed significant aspects of the disease and the devils' responses to it. These include the cause and pathogenesis of DFTD, the immune response of the devils and the immune evasion mechanisms of the tumor, the transmission patterns of DFTD, and the impacts of DFTD on the ecosystem. This review aims to collate this information and put it into the context of conservation strategies designed to mitigate the impacts of DFTD on the devil and the Tasmanian ecosystem.
Collapse
Affiliation(s)
- R J Pye
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Tasmania, Australia
| | - G M Woods
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Tasmania, Australia
| | - A Kreiss
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Tasmania, Australia
| |
Collapse
|
8
|
Medullomyoblastoma treated with craniospinal radiation and adjuvant chemotherapy: Report of 4 cases and review of the literature. J Egypt Natl Canc Inst 2015; 27:109-11. [DOI: 10.1016/j.jnci.2015.03.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2015] [Revised: 03/19/2015] [Accepted: 03/20/2015] [Indexed: 11/21/2022] Open
|
9
|
Abstract
Medulloblastoma is the most common malignant brain tumor of childhood. Although there is now long-term survival or cure for the majority of children, the survivors bear a significant burden of complications due, at least in part, to the intense therapies given to ensure eradication of the tumor. Significant efforts have been made over the years to be able to distinguish between patients who do and do not need intensive therapies. This review summarizes the history and current state of clinical risk stratification, pathologic diagnosis and genetics. Recent developments in correlation between genetics and pathology, genome-wide association studies and the biology of medulloblastoma metastasis are discussed in detail. The current state of clinical treatment trials are reviewed and placed into the perspective of potential novel therapies in the near term.
Collapse
Affiliation(s)
- Donya Aref
- University Health Network Pathology, Arthur & Sonia Labatt Brain Tumour Research Centre, Department of Laboratory Medicine & Pathobiology, Toronto, ON, Canada
| | | |
Collapse
|
10
|
Clement J, Varlotto J, Rybka W, Frauenhoffer E, Drabick JJ. Unusual case of recurrent extraneural metastatic medulloblastoma in a young adult: durable complete remission with Ewing sarcoma chemotherapy regimen and consolidation with autologous bone marrow transplantation and local radiation. J Clin Oncol 2013; 31:e316-9. [PMID: 23715566 DOI: 10.1200/jco.2012.42.6700] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Affiliation(s)
- Jincy Clement
- Department of Internal Medicine, Penn State Milton S. Hershey Medical Center, Hershey, PA, USA.
| | | | | | | | | |
Collapse
|
11
|
Lannering B, Rutkowski S, Doz F, Pizer B, Gustafsson G, Navajas A, Massimino M, Reddingius R, Benesch M, Carrie C, Taylor R, Gandola L, Björk-Eriksson T, Giralt J, Oldenburger F, Pietsch T, Figarella-Branger D, Robson K, Forni M, Clifford SC, Warmuth-Metz M, von Hoff K, Faldum A, Mosseri V, Kortmann R. Hyperfractionated Versus Conventional Radiotherapy Followed by Chemotherapy in Standard-Risk Medulloblastoma: Results From the Randomized Multicenter HIT-SIOP PNET 4 Trial. J Clin Oncol 2012; 30:3187-93. [DOI: 10.1200/jco.2011.39.8719] [Citation(s) in RCA: 223] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Purpose To compare event-free survival (EFS), overall survival (OS), pattern of relapse, and hearing loss in children with standard-risk medulloblastoma treated by postoperative hyperfractionated or conventionally fractionated radiotherapy followed by maintenance chemotherapy. Patients and Methods In all, 340 children age 4 to 21 years from 122 European centers were postoperatively staged and randomly assigned to treatment with hyperfractionated radiotherapy (HFRT) or standard (conventional) fractionated radiotherapy (STRT) followed by a common chemotherapy regimen consisting of eight cycles of cisplatin, lomustine, and vincristine. Results After a median follow-up of 4.8 years (range, 0.1 to 8.3 years), survival rates were not significantly different between the two treatment arms: 5-year EFS was 77% ± 4% in the STRT group and 78% ± 4% in the HFRT group; corresponding 5-year OS was 87% ± 3% and 85% ± 3%, respectively. A postoperative residual tumor of more than 1.5 cm2 was the strongest negative prognostic factor. EFS of children with all reference assessments and no large residual tumor was 82% ± 2% at 5 years. Patients with a delay of more than 7 weeks to the start of RT had a worse prognosis. Severe hearing loss was not significantly different for the two treatment arms at follow-up. Conclusion In this large randomized European study, which enrolled patients with standard-risk medulloblastoma from more than 100 centers, excellent survival rates were achieved in patients without a large postoperative residual tumor and without RT treatment delays. EFS and OS for HFRT was not superior to STRT, which therefore remains standard of care in this disease.
Collapse
Affiliation(s)
- Birgitta Lannering
- Birgitta Lannering and Thomas Björk-Eriksson, University of Gothenburg, Göteborg; Göran Gustafsson, Karolinska Institute, Stockholm, Sweden; Stefan Rutkowski and Katja von Hoff, University Medical Center Hamburg-Eppendorf, Hamburg; Torsten Pietsch, University of Bonn, Bonn; Monica Warmuth-Metz, University of Wuerzburg, Wuerzburg; Andreas Faldum, Institute for Biometry and Clinical Research, University of Munster, Munster; Rolf Kortmann, University of Leipzig, Leipzig, Germany; Francois Doz, Institut
| | - Stefan Rutkowski
- Birgitta Lannering and Thomas Björk-Eriksson, University of Gothenburg, Göteborg; Göran Gustafsson, Karolinska Institute, Stockholm, Sweden; Stefan Rutkowski and Katja von Hoff, University Medical Center Hamburg-Eppendorf, Hamburg; Torsten Pietsch, University of Bonn, Bonn; Monica Warmuth-Metz, University of Wuerzburg, Wuerzburg; Andreas Faldum, Institute for Biometry and Clinical Research, University of Munster, Munster; Rolf Kortmann, University of Leipzig, Leipzig, Germany; Francois Doz, Institut
| | - Francois Doz
- Birgitta Lannering and Thomas Björk-Eriksson, University of Gothenburg, Göteborg; Göran Gustafsson, Karolinska Institute, Stockholm, Sweden; Stefan Rutkowski and Katja von Hoff, University Medical Center Hamburg-Eppendorf, Hamburg; Torsten Pietsch, University of Bonn, Bonn; Monica Warmuth-Metz, University of Wuerzburg, Wuerzburg; Andreas Faldum, Institute for Biometry and Clinical Research, University of Munster, Munster; Rolf Kortmann, University of Leipzig, Leipzig, Germany; Francois Doz, Institut
| | - Barry Pizer
- Birgitta Lannering and Thomas Björk-Eriksson, University of Gothenburg, Göteborg; Göran Gustafsson, Karolinska Institute, Stockholm, Sweden; Stefan Rutkowski and Katja von Hoff, University Medical Center Hamburg-Eppendorf, Hamburg; Torsten Pietsch, University of Bonn, Bonn; Monica Warmuth-Metz, University of Wuerzburg, Wuerzburg; Andreas Faldum, Institute for Biometry and Clinical Research, University of Munster, Munster; Rolf Kortmann, University of Leipzig, Leipzig, Germany; Francois Doz, Institut
| | - Göran Gustafsson
- Birgitta Lannering and Thomas Björk-Eriksson, University of Gothenburg, Göteborg; Göran Gustafsson, Karolinska Institute, Stockholm, Sweden; Stefan Rutkowski and Katja von Hoff, University Medical Center Hamburg-Eppendorf, Hamburg; Torsten Pietsch, University of Bonn, Bonn; Monica Warmuth-Metz, University of Wuerzburg, Wuerzburg; Andreas Faldum, Institute for Biometry and Clinical Research, University of Munster, Munster; Rolf Kortmann, University of Leipzig, Leipzig, Germany; Francois Doz, Institut
| | - Aurora Navajas
- Birgitta Lannering and Thomas Björk-Eriksson, University of Gothenburg, Göteborg; Göran Gustafsson, Karolinska Institute, Stockholm, Sweden; Stefan Rutkowski and Katja von Hoff, University Medical Center Hamburg-Eppendorf, Hamburg; Torsten Pietsch, University of Bonn, Bonn; Monica Warmuth-Metz, University of Wuerzburg, Wuerzburg; Andreas Faldum, Institute for Biometry and Clinical Research, University of Munster, Munster; Rolf Kortmann, University of Leipzig, Leipzig, Germany; Francois Doz, Institut
| | - Maura Massimino
- Birgitta Lannering and Thomas Björk-Eriksson, University of Gothenburg, Göteborg; Göran Gustafsson, Karolinska Institute, Stockholm, Sweden; Stefan Rutkowski and Katja von Hoff, University Medical Center Hamburg-Eppendorf, Hamburg; Torsten Pietsch, University of Bonn, Bonn; Monica Warmuth-Metz, University of Wuerzburg, Wuerzburg; Andreas Faldum, Institute for Biometry and Clinical Research, University of Munster, Munster; Rolf Kortmann, University of Leipzig, Leipzig, Germany; Francois Doz, Institut
| | - Roel Reddingius
- Birgitta Lannering and Thomas Björk-Eriksson, University of Gothenburg, Göteborg; Göran Gustafsson, Karolinska Institute, Stockholm, Sweden; Stefan Rutkowski and Katja von Hoff, University Medical Center Hamburg-Eppendorf, Hamburg; Torsten Pietsch, University of Bonn, Bonn; Monica Warmuth-Metz, University of Wuerzburg, Wuerzburg; Andreas Faldum, Institute for Biometry and Clinical Research, University of Munster, Munster; Rolf Kortmann, University of Leipzig, Leipzig, Germany; Francois Doz, Institut
| | - Martin Benesch
- Birgitta Lannering and Thomas Björk-Eriksson, University of Gothenburg, Göteborg; Göran Gustafsson, Karolinska Institute, Stockholm, Sweden; Stefan Rutkowski and Katja von Hoff, University Medical Center Hamburg-Eppendorf, Hamburg; Torsten Pietsch, University of Bonn, Bonn; Monica Warmuth-Metz, University of Wuerzburg, Wuerzburg; Andreas Faldum, Institute for Biometry and Clinical Research, University of Munster, Munster; Rolf Kortmann, University of Leipzig, Leipzig, Germany; Francois Doz, Institut
| | - Christian Carrie
- Birgitta Lannering and Thomas Björk-Eriksson, University of Gothenburg, Göteborg; Göran Gustafsson, Karolinska Institute, Stockholm, Sweden; Stefan Rutkowski and Katja von Hoff, University Medical Center Hamburg-Eppendorf, Hamburg; Torsten Pietsch, University of Bonn, Bonn; Monica Warmuth-Metz, University of Wuerzburg, Wuerzburg; Andreas Faldum, Institute for Biometry and Clinical Research, University of Munster, Munster; Rolf Kortmann, University of Leipzig, Leipzig, Germany; Francois Doz, Institut
| | - Roger Taylor
- Birgitta Lannering and Thomas Björk-Eriksson, University of Gothenburg, Göteborg; Göran Gustafsson, Karolinska Institute, Stockholm, Sweden; Stefan Rutkowski and Katja von Hoff, University Medical Center Hamburg-Eppendorf, Hamburg; Torsten Pietsch, University of Bonn, Bonn; Monica Warmuth-Metz, University of Wuerzburg, Wuerzburg; Andreas Faldum, Institute for Biometry and Clinical Research, University of Munster, Munster; Rolf Kortmann, University of Leipzig, Leipzig, Germany; Francois Doz, Institut
| | - Lorenza Gandola
- Birgitta Lannering and Thomas Björk-Eriksson, University of Gothenburg, Göteborg; Göran Gustafsson, Karolinska Institute, Stockholm, Sweden; Stefan Rutkowski and Katja von Hoff, University Medical Center Hamburg-Eppendorf, Hamburg; Torsten Pietsch, University of Bonn, Bonn; Monica Warmuth-Metz, University of Wuerzburg, Wuerzburg; Andreas Faldum, Institute for Biometry and Clinical Research, University of Munster, Munster; Rolf Kortmann, University of Leipzig, Leipzig, Germany; Francois Doz, Institut
| | - Thomas Björk-Eriksson
- Birgitta Lannering and Thomas Björk-Eriksson, University of Gothenburg, Göteborg; Göran Gustafsson, Karolinska Institute, Stockholm, Sweden; Stefan Rutkowski and Katja von Hoff, University Medical Center Hamburg-Eppendorf, Hamburg; Torsten Pietsch, University of Bonn, Bonn; Monica Warmuth-Metz, University of Wuerzburg, Wuerzburg; Andreas Faldum, Institute for Biometry and Clinical Research, University of Munster, Munster; Rolf Kortmann, University of Leipzig, Leipzig, Germany; Francois Doz, Institut
| | - Jordi Giralt
- Birgitta Lannering and Thomas Björk-Eriksson, University of Gothenburg, Göteborg; Göran Gustafsson, Karolinska Institute, Stockholm, Sweden; Stefan Rutkowski and Katja von Hoff, University Medical Center Hamburg-Eppendorf, Hamburg; Torsten Pietsch, University of Bonn, Bonn; Monica Warmuth-Metz, University of Wuerzburg, Wuerzburg; Andreas Faldum, Institute for Biometry and Clinical Research, University of Munster, Munster; Rolf Kortmann, University of Leipzig, Leipzig, Germany; Francois Doz, Institut
| | - Foppe Oldenburger
- Birgitta Lannering and Thomas Björk-Eriksson, University of Gothenburg, Göteborg; Göran Gustafsson, Karolinska Institute, Stockholm, Sweden; Stefan Rutkowski and Katja von Hoff, University Medical Center Hamburg-Eppendorf, Hamburg; Torsten Pietsch, University of Bonn, Bonn; Monica Warmuth-Metz, University of Wuerzburg, Wuerzburg; Andreas Faldum, Institute for Biometry and Clinical Research, University of Munster, Munster; Rolf Kortmann, University of Leipzig, Leipzig, Germany; Francois Doz, Institut
| | - Torsten Pietsch
- Birgitta Lannering and Thomas Björk-Eriksson, University of Gothenburg, Göteborg; Göran Gustafsson, Karolinska Institute, Stockholm, Sweden; Stefan Rutkowski and Katja von Hoff, University Medical Center Hamburg-Eppendorf, Hamburg; Torsten Pietsch, University of Bonn, Bonn; Monica Warmuth-Metz, University of Wuerzburg, Wuerzburg; Andreas Faldum, Institute for Biometry and Clinical Research, University of Munster, Munster; Rolf Kortmann, University of Leipzig, Leipzig, Germany; Francois Doz, Institut
| | - Dominique Figarella-Branger
- Birgitta Lannering and Thomas Björk-Eriksson, University of Gothenburg, Göteborg; Göran Gustafsson, Karolinska Institute, Stockholm, Sweden; Stefan Rutkowski and Katja von Hoff, University Medical Center Hamburg-Eppendorf, Hamburg; Torsten Pietsch, University of Bonn, Bonn; Monica Warmuth-Metz, University of Wuerzburg, Wuerzburg; Andreas Faldum, Institute for Biometry and Clinical Research, University of Munster, Munster; Rolf Kortmann, University of Leipzig, Leipzig, Germany; Francois Doz, Institut
| | - Keith Robson
- Birgitta Lannering and Thomas Björk-Eriksson, University of Gothenburg, Göteborg; Göran Gustafsson, Karolinska Institute, Stockholm, Sweden; Stefan Rutkowski and Katja von Hoff, University Medical Center Hamburg-Eppendorf, Hamburg; Torsten Pietsch, University of Bonn, Bonn; Monica Warmuth-Metz, University of Wuerzburg, Wuerzburg; Andreas Faldum, Institute for Biometry and Clinical Research, University of Munster, Munster; Rolf Kortmann, University of Leipzig, Leipzig, Germany; Francois Doz, Institut
| | - Marco Forni
- Birgitta Lannering and Thomas Björk-Eriksson, University of Gothenburg, Göteborg; Göran Gustafsson, Karolinska Institute, Stockholm, Sweden; Stefan Rutkowski and Katja von Hoff, University Medical Center Hamburg-Eppendorf, Hamburg; Torsten Pietsch, University of Bonn, Bonn; Monica Warmuth-Metz, University of Wuerzburg, Wuerzburg; Andreas Faldum, Institute for Biometry and Clinical Research, University of Munster, Munster; Rolf Kortmann, University of Leipzig, Leipzig, Germany; Francois Doz, Institut
| | - Steven C. Clifford
- Birgitta Lannering and Thomas Björk-Eriksson, University of Gothenburg, Göteborg; Göran Gustafsson, Karolinska Institute, Stockholm, Sweden; Stefan Rutkowski and Katja von Hoff, University Medical Center Hamburg-Eppendorf, Hamburg; Torsten Pietsch, University of Bonn, Bonn; Monica Warmuth-Metz, University of Wuerzburg, Wuerzburg; Andreas Faldum, Institute for Biometry and Clinical Research, University of Munster, Munster; Rolf Kortmann, University of Leipzig, Leipzig, Germany; Francois Doz, Institut
| | - Monica Warmuth-Metz
- Birgitta Lannering and Thomas Björk-Eriksson, University of Gothenburg, Göteborg; Göran Gustafsson, Karolinska Institute, Stockholm, Sweden; Stefan Rutkowski and Katja von Hoff, University Medical Center Hamburg-Eppendorf, Hamburg; Torsten Pietsch, University of Bonn, Bonn; Monica Warmuth-Metz, University of Wuerzburg, Wuerzburg; Andreas Faldum, Institute for Biometry and Clinical Research, University of Munster, Munster; Rolf Kortmann, University of Leipzig, Leipzig, Germany; Francois Doz, Institut
| | - Katja von Hoff
- Birgitta Lannering and Thomas Björk-Eriksson, University of Gothenburg, Göteborg; Göran Gustafsson, Karolinska Institute, Stockholm, Sweden; Stefan Rutkowski and Katja von Hoff, University Medical Center Hamburg-Eppendorf, Hamburg; Torsten Pietsch, University of Bonn, Bonn; Monica Warmuth-Metz, University of Wuerzburg, Wuerzburg; Andreas Faldum, Institute for Biometry and Clinical Research, University of Munster, Munster; Rolf Kortmann, University of Leipzig, Leipzig, Germany; Francois Doz, Institut
| | - Andreas Faldum
- Birgitta Lannering and Thomas Björk-Eriksson, University of Gothenburg, Göteborg; Göran Gustafsson, Karolinska Institute, Stockholm, Sweden; Stefan Rutkowski and Katja von Hoff, University Medical Center Hamburg-Eppendorf, Hamburg; Torsten Pietsch, University of Bonn, Bonn; Monica Warmuth-Metz, University of Wuerzburg, Wuerzburg; Andreas Faldum, Institute for Biometry and Clinical Research, University of Munster, Munster; Rolf Kortmann, University of Leipzig, Leipzig, Germany; Francois Doz, Institut
| | - Véronique Mosseri
- Birgitta Lannering and Thomas Björk-Eriksson, University of Gothenburg, Göteborg; Göran Gustafsson, Karolinska Institute, Stockholm, Sweden; Stefan Rutkowski and Katja von Hoff, University Medical Center Hamburg-Eppendorf, Hamburg; Torsten Pietsch, University of Bonn, Bonn; Monica Warmuth-Metz, University of Wuerzburg, Wuerzburg; Andreas Faldum, Institute for Biometry and Clinical Research, University of Munster, Munster; Rolf Kortmann, University of Leipzig, Leipzig, Germany; Francois Doz, Institut
| | - Rolf Kortmann
- Birgitta Lannering and Thomas Björk-Eriksson, University of Gothenburg, Göteborg; Göran Gustafsson, Karolinska Institute, Stockholm, Sweden; Stefan Rutkowski and Katja von Hoff, University Medical Center Hamburg-Eppendorf, Hamburg; Torsten Pietsch, University of Bonn, Bonn; Monica Warmuth-Metz, University of Wuerzburg, Wuerzburg; Andreas Faldum, Institute for Biometry and Clinical Research, University of Munster, Munster; Rolf Kortmann, University of Leipzig, Leipzig, Germany; Francois Doz, Institut
| |
Collapse
|
12
|
Markant SL, Wechsler-Reya RJ. Personalized mice: modelling the molecular heterogeneity of medulloblastoma. Neuropathol Appl Neurobiol 2012; 38:228-40. [DOI: 10.1111/j.1365-2990.2011.01235.x] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
13
|
Bien-Willner GA, López-Terrada D, Bhattacharjee MB, Patel KU, Stankiewicz P, Lupski JR, Pfeifer JD, Perry A. Early recurrence in standard-risk medulloblastoma patients with the common idic(17)(p11.2) rearrangement. Neuro Oncol 2012; 14:831-40. [PMID: 22573308 DOI: 10.1093/neuonc/nos086] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Medulloblastoma is diagnosed histologically; treatment depends on staging and age of onset. Whereas clinical factors identify a standard- and a high-risk population, these findings cannot differentiate which standard-risk patients will relapse and die. Outcome is thought to be influenced by tumor subtype and molecular alterations. Poor prognosis has been associated with isochromosome (i)17q in some but not all studies. In most instances, molecular investigations document that i17q is not a true isochromosome but rather an isodicentric chromosome, idic(17)(p11.2), with rearrangement breakpoints mapping within the REPA/REPB region on 17p11.2. This study explores the clinical utility of testing for idic(17)(p11.2) rearrangements using an assay based on fluorescent in situ hybridization (FISH). This test was applied to 58 consecutive standard- and high-risk medulloblastomas with a 5-year minimum of clinical follow-up. The presence of i17q (ie, including cases not involving the common breakpoint), idic(17)(p11.2), and histologic subtype was correlated with clinical outcome. Overall survival (OS) and disease-free survival (DFS) were consistent with literature reports. Fourteen patients (25%) had i17q, with 10 (18%) involving the common isodicentric rearrangement. The presence of i17q was associated with a poor prognosis. OS and DFS were poor in all cases with anaplasia (4), unresectable disease (7), and metastases at presentation (10); however, patients with standard-risk tumors fared better. Of these 44 cases, tumors with idic(17)(p11.2) were associated with significantly worse patient outcomes and shorter mean DFS. FISH detection of idic(17)(p11.2) may be useful for risk stratification in standard-risk patients. The presence of this abnormal chromosome is associated with early recurrence of medulloblastoma.
Collapse
|
14
|
Abstract
Myc proteins are often deregulated in human brain tumors, especially in embryonal tumors that affect children. Many observations have shown how alterations of these pleiotropic Myc transcription factors provide initiation, maintenance, or progression of tumors. This review will focus on the role of Myc family members (particularly c-myc and Mycn) in tumors like medulloblastoma and glioma and will further discuss how to target stabilization of these proteins for future brain tumor therapies.
Collapse
Affiliation(s)
- Fredrik J Swartling
- Uppsala University, Department of Immunology, Genetics, and Pathology, Rudbeck Laboratory, Uppsala, Sweden.
| |
Collapse
|
15
|
Pei Y, Moore CE, Wang J, Tewari AK, Eroshkin A, Cho YJ, Witt H, Korshunov A, Read TA, Sun JL, Schmitt EM, Miller CR, Buckley AF, McLendon RE, Westbrook TF, Northcott PA, Taylor MD, Pfister SM, Febbo PG, Wechsler-Reya RJ. An animal model of MYC-driven medulloblastoma. Cancer Cell 2012; 21:155-67. [PMID: 22340590 PMCID: PMC3285431 DOI: 10.1016/j.ccr.2011.12.021] [Citation(s) in RCA: 237] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2011] [Revised: 11/21/2011] [Accepted: 12/22/2011] [Indexed: 12/11/2022]
Abstract
Medulloblastoma (MB) is the most common malignant brain tumor in children. Patients whose tumors exhibit overexpression or amplification of the MYC oncogene (c-MYC) usually have an extremely poor prognosis, but there are no animal models of this subtype of the disease. Here, we show that cerebellar stem cells expressing Myc and mutant Trp53 (p53) generate aggressive tumors following orthotopic transplantation. These tumors consist of large, pleiomorphic cells and resemble human MYC-driven MB at a molecular level. Notably, antagonists of PI3K/mTOR signaling, but not Hedgehog signaling, inhibit growth of tumor cells. These findings suggest that cerebellar stem cells can give rise to MYC-driven MB and identify a novel model that can be used to test therapies for this devastating disease.
Collapse
Affiliation(s)
- Yanxin Pei
- Tumor Development Program, Sanford-Burnham Medical Research Institute, La Jolla, CA
| | - Colin E. Moore
- Tumor Development Program, Sanford-Burnham Medical Research Institute, La Jolla, CA
| | - Jun Wang
- Tumor Development Program, Sanford-Burnham Medical Research Institute, La Jolla, CA
| | - Alok K. Tewari
- Department of Medicine and Helen Diller Family Comprehensive Cancer Center, UCSF, San Francisco, CA
| | - Alexey Eroshkin
- Bioinformatics Shared Resource, Sanford-Burnham Medical Research Institute, La Jolla, CA
| | - Yoon-Jae Cho
- Stanford University School of Medicine, Stanford, CA
| | - Hendrik Witt
- German Cancer Research Center and University of Heidelberg, Heidelberg, Germany
| | - Andrey Korshunov
- German Cancer Research Center and University of Heidelberg, Heidelberg, Germany
| | - Tracy-Ann Read
- Department of Neurosurgery, Emory University School of Medicine, Atlanta, GA
| | - Julia L. Sun
- Department of Pharmacology & Cancer Biology, Duke University Medical Center, Durham, NC
| | - Earlene M. Schmitt
- Verna & Marrs McLean Department of Biochemistry & Molecular Biology, and Department of Molecular & Human Genetics, Baylor College of Medicine, Houston, TX
| | - C. Ryan Miller
- Department of Pathology and Laboratory Medicine, UNC, Chapel Hill, NC
| | - Anne F. Buckley
- Department of Pathology, Duke University Medical Center, Durham, NC
| | | | - Thomas F. Westbrook
- Verna & Marrs McLean Department of Biochemistry & Molecular Biology, and Department of Molecular & Human Genetics, Baylor College of Medicine, Houston, TX
- Department of Pediatrics and Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX
| | - Paul A. Northcott
- Hospital for Sick Children and University of Toronto, Toronto, Ontario, Canada
| | - Michael D. Taylor
- Hospital for Sick Children and University of Toronto, Toronto, Ontario, Canada
| | - Stefan M. Pfister
- German Cancer Research Center and University of Heidelberg, Heidelberg, Germany
| | - Phillip G. Febbo
- Department of Medicine and Helen Diller Family Comprehensive Cancer Center, UCSF, San Francisco, CA
| | - Robert J. Wechsler-Reya
- Tumor Development Program, Sanford-Burnham Medical Research Institute, La Jolla, CA
- Department of Pharmacology & Cancer Biology, Duke University Medical Center, Durham, NC
- To whom correspondence should be addressed: Tumor Development Program, Sanford-Burnham Medical Research Institute, 10901, North Torrey Pines Road, La Jolla, CA 92037,
| |
Collapse
|
16
|
McLendon RE, Adekunle A, Rajaram V, Koçak M, Blaney SM. Embryonal central nervous system neoplasms arising in infants and young children: a pediatric brain tumor consortium study. Arch Pathol Lab Med 2011; 135:984-93. [PMID: 21809989 PMCID: PMC3752842 DOI: 10.5858/2010-0515-oar1] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
CONTEXT Medulloblastomas (MBs) and atypical teratoid/rhabdoid tumors (AT/RTs) arising in infants and children can be difficult to distinguish; however, histologic characterization is prognostically important. OBJECTIVE To determine histologic and phenotypic markers associated with utility with progression-free survival (PFS) and overall survival (OS) in children younger than 3 years with MBs and AT/RTs. DESIGN We undertook a histologic and immunophenotypic study of MBs and AT/RTs arising in infants and children younger than 3 years treated in a Pediatric Brain Tumor Consortium study. The 41 girls and 55 boys ranged in age from 2 to 36 months at enrollment. These infants and children exhibited 51 MBs, 26 AT/RTs, and 24 other tumors (not further studied). Median follow-up of the patients was 17.2 months from diagnosis (range: 1.4-93 months). RESULTS Infants and children with AT/RT exhibited a statistically significant shorter PFS and OS when compared to infants and children with MBs (both P < .001). A lack of nuclear BAF47 immunohistochemical reactivity proved reliable in identifying AT/RTs. Among MBs, our data suggest an association of nodularity and prolonged PFS and OS, which must be independently confirmed. Anaplasia correlated with OTX2 reactivity and both OTX2 and moderate to severe anaplasia correlated with PFS but not with OS. CONCLUSION Distinguishing AT/RT from MBs is clinically important. For expert neuropathologists, the diagnoses of AT/RT and MB can be reliably made from hematoxylin-eosin stains in the vast majority of cases. However certain rare small cell variants of AT/RT can be confused with MB. We also found that immunohistochemical reactivity for BAF47 is clinically useful in distinguishing MBs from AT/RTs and for identifying certain small cell AT/RTs. Among MBs, nodularity may be an important prognostic factor for improved PFS and OS in infants and children.
Collapse
Affiliation(s)
- Roger E McLendon
- Department of Pathology, Duke University Medical Center, Durham, North Carolina 27710, USA.
| | | | | | | | | |
Collapse
|
17
|
Chan MD, Attia A, Tatter SB, Lesser G, Zapadka ME, Mott RT, Carter A, McMullen KP, Shaw EG, Ellis TE. Radiation-induced adult medulloblastoma: a two-case report and review of the literature. J Neurooncol 2010; 103:745-9. [PMID: 20936325 DOI: 10.1007/s11060-010-0426-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2010] [Accepted: 09/25/2010] [Indexed: 10/19/2022]
Abstract
Radiation-induced medulloblastoma is an exceedingly rare phenomenon for which treatment standards have not been established. The literature suggests that these tumors are high grade with aggressive behavior. We report two cases of radiation-induced medulloblastoma which have been treated with full dose re-irradiation with curative intent. In both cases, treatment toxicity and tumor progression proved to be insurmountable obstacles. Further reports are necessary in order to fully characterize this clinical entity so that more effective therapies may be sought.
Collapse
Affiliation(s)
- Michael D Chan
- Department of Radiation Oncology, Wake Forest University Health Sciences-Baptist Medical Center, 1 Medical Center Blvd, Winston-Salem, NC 27103, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Zhou L, Picard D, Ra YS, Li M, Northcott PA, Hu Y, Stearns D, Hawkins C, Taylor MD, Rutka J, Der SD, Huang A. Silencing of thrombospondin-1 is critical for myc-induced metastatic phenotypes in medulloblastoma. Cancer Res 2010; 70:8199-210. [PMID: 20876797 DOI: 10.1158/0008-5472.can-09-4562] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Mechanisms by which c-Myc (Myc) amplification confers aggressive medulloblastoma phenotypes are poorly defined. Here, we show using orthotopic models that high Myc expression promotes cell migration/invasion and induces metastatic tumors, which recapitulate aggressive histologic features of Myc-amplified primary human medulloblastoma. Using ChIP-chip analysis, we identified cell migration and adhesion genes, including Tsp-1/THBS1, ING4, PVRL3, and PPAP2B, as Myc-bound loci in medulloblastoma cells. Expression of Tsp-1 was most consistently and robustly diminished in medulloblastoma cell lines and primary human tumors with high Myc expression (n = 101, P = 0.032). Strikingly, stable Tsp-1 expression significantly attenuated in vitro transformation and invasive/migratory properties of high Myc-expressing medulloblastoma cells without altering cell proliferation, whereas RNA interference-mediated Myc knockdown was consistently accompanied by increased Tsp-1 levels and reduced cell migration and invasion in medulloblastoma cells. Chromatin immunoprecipitation (ChIP) assays revealed colocalization of Myc and obligate partner Max and correlated diminished RNA polymerase II occupancy (∼3-fold decrease, P < 0.01) with increased Myc binding at a core Tsp-1 promoter. Reporter gene and/or gel shift assays confirmed direct repression of Tsp-1 transcription by Myc and also identified JPO2, a Myc interactor associated with metastatic medulloblastoma, as a cofactor in Myc-mediated Tsp-1 repression. These findings indicate the Myc-regulatory network targets Tsp-1 via multiple mechanisms in medulloblastoma transformation, and highlight a novel critical role for Tsp-1 in Myc-mediated aggressive medulloblastoma phenotypes.
Collapse
Affiliation(s)
- Limei Zhou
- Sonia and Arthur Labatt Brain Tumor Research Centre, Hospital for Sick Children, University Avenue, Toronto, Ontario, Canada M5G 1X8
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Childhood medulloblastoma: novel approaches to the classification of a heterogeneous disease. Acta Neuropathol 2010; 120:305-16. [PMID: 20652577 DOI: 10.1007/s00401-010-0726-6] [Citation(s) in RCA: 77] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2010] [Revised: 07/12/2010] [Accepted: 07/12/2010] [Indexed: 10/19/2022]
Abstract
Increasing recognition of pediatric medulloblastoma as a heterogeneous disease, with histopathological and molecular variants that have distinct biological behaviors, will impact how the disease is classified and treated. A combination of clinicopathological evaluation and assays based on molecular subgroups of disease will allow stratification of patients into risk groups and a more tailored approach to therapy. Patients with low-risk disease could be treated with de-escalated adjuvant therapy to maximize cure while reducing long-term adverse effects, and novel therapies could be sought for patients with high-risk disease. My review encompasses a brief overview of the clinical landscape, the current World Health Organization (WHO) classification of medulloblastoma, the status of molecular subgroups, and how potential stratification schemes might impact pathologists and their practice.
Collapse
|
20
|
Swartling FJ, Grimmer MR, Hackett CS, Northcott PA, Fan QW, Goldenberg DD, Lau J, Masic S, Nguyen K, Yakovenko S, Zhe XN, Gilmer HCF, Collins R, Nagaoka M, Phillips JJ, Jenkins RB, Tihan T, Vandenberg SR, James CD, Tanaka K, Taylor MD, Weiss WA, Chesler L. Pleiotropic role for MYCN in medulloblastoma. Genes Dev 2010; 24:1059-72. [PMID: 20478998 DOI: 10.1101/gad.1907510] [Citation(s) in RCA: 138] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Medulloblastoma (MB) is the most common malignant brain tumor of childhood. Sonic Hedgehog (SHH) signaling drives a minority of MB, correlating with desmoplastic pathology and favorable outcome. The majority, however, arises independently of SHH and displays classic or large cell anaplastic (LCA) pathology and poor prognosis. To identify common signaling abnormalities, we profiled mRNA, demonstrating misexpression of MYCN in the majority of human MB and negligible expression in normal cerebella. We clarified a role in pathogenesis by targeting MYCN (and luciferase) to cerebella of transgenic mice. MYCN-driven MB showed either classic or LCA pathologies, with Shh signaling activated in approximately 5% of tumors, demonstrating that MYCN can drive MB independently of Shh. MB arose at high penetrance, consistent with a role for MYCN in initiation. Tumor burden correlated with bioluminescence, with rare metastatic spread to the leptomeninges, suggesting roles for MYCN in both progression and metastasis. Transient pharmacological down-regulation of MYCN led to both clearance and senescence of tumor cells, and improved survival. Targeted expression of MYCN thus contributes to initiation, progression, and maintenance of MB, suggesting a central role for MYCN in pathogenesis.
Collapse
Affiliation(s)
- Fredrik J Swartling
- University of California at San Francisco, San Francisco, California 94158, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Srikantha U, Balasubramaniam A, Santosh V, Somanna S, Bhagavatula ID, Ashwathnarayana CB. Recurrence in medulloblastoma – Influence of clinical, histological and immunohistochemical factors. Br J Neurosurg 2010; 24:280-8. [DOI: 10.3109/02688691003660558] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
|
22
|
Abstract
Medulloblastoma, a primitive neuro-ectodermal tumour that arises in the posterior fossa, is the most common malignant brain tumour occurring in childhood. Over the past half century, the long-term survival for children with medulloblastoma has improved remarkably from a certain fatal diagnosis to a cancer that is often curable. Although overall survival for children with non-disseminated and non-anaplastic medulloblastoma can approach 80%, the current multidisciplinary therapeutic approach is not without long-term sequelae. Chemotherapy has improved the long-term survival and allowed for reductions in the amount of radiation given, thereby reducing some of the long-term toxicities. In this review, we describe the current understanding of the basic biology of medulloblastoma and report on the current active chemotherapeutic agents utilized in medulloblastoma therapy. Ultimately, our understanding of the basic biology of medulloblastoma may lead to further advances in therapy by providing targets that are more specific and potentially less toxic.
Collapse
Affiliation(s)
- Laura J Klesse
- Division of Pediatric Hematology-Oncology, Department of Pediatrics, University of Texas Southwestern Medical Center at Dallas, Dallas, Texas 75390-9063, USA.
| | | |
Collapse
|
23
|
Raabe EH, Eberhart CG. High-risk medulloblastoma: does c-myc amplification overrule histopathology? Pediatr Blood Cancer 2010; 54:344-5. [PMID: 20063409 PMCID: PMC4517433 DOI: 10.1002/pbc.22398] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Affiliation(s)
- Eric H. Raabe
- Division of Pediatric Oncology, Johns Hopkins University School of
Medicine, Baltimore, Maryland,Correspondence to: Eric H. Raabe, Division of Pediatric
Oncology, Johns Hopkins Hospital, CMSC 800, 600 N. Wolfe St., Baltimore, MD
21287.
| | - Charles G. Eberhart
- Division of Neuropathology, Johns Hopkins University School of
Medicine, Baltimore, Maryland,Division of Ophthalmic Pathology, Johns Hopkins University School of
Medicine, Baltimore, Maryland
| |
Collapse
|
24
|
Adamson DC, Shi Q, Wortham M, Northcott PA, Di C, Duncan CG, Li J, McLendon RE, Bigner DD, Taylor MD, Yan H. OTX2 is critical for the maintenance and progression of Shh-independent medulloblastomas. Cancer Res 2009; 70:181-91. [PMID: 20028867 DOI: 10.1158/0008-5472.can-09-2331] [Citation(s) in RCA: 89] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
OTX2 is a developmentally regulated transcription factor involved in early morphogenesis of the central nervous system. This gene is amplified and overexpressed in medulloblastoma cell lines, but the nature and extent of its genetic alterations in primary tumors have not been evaluated. Analysis of a large cohort of primary medulloblastomas revealed frequent focal copy number gain of a region minimally containing OTX2 as a single gene. OTX2 copy number gain was restricted to tumor subtypes that did not express a molecular signature of Wnt or Shh pathway activation. FISH analysis revealed copy number gain in a subset of cells within medulloblastoma samples, suggesting a late event in tumor progression. Gain of OTX2 copy number was associated with the presence of anaplastic histologic features and shorter survival in medulloblastoma patients. In support of a functional role, ectopic OTX2 expression enhanced proliferation and tumorigenicity of immortalized primary cells, whereas OTX2 knockdown in medulloblastoma cells prolonged the survival of animals bearing xenograft tumors. Mechanistic investigations revealed upregulation of MYC as a potential mechanism whereby OTX2 promotes tumor progression. Our findings define OTX2 as an important oncogenic driver in medulloblastoma.
Collapse
Affiliation(s)
- David C Adamson
- Department of Surgery, The Pediatric Brain Tumor Foundation Institute, and The Preston Robert Tisch Brain Tumor Center at Duke, Duke University Medical Center, Veterans Affairs Medical Center, Durham, North Carolina 27710, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Medulloblastomas: a correlative study of MIB-1 proliferation index along with expression of c-Myc, ERBB2, and anti-apoptotic proteins along with histological typing and clinical outcome. Childs Nerv Syst 2009; 25:825-35. [PMID: 19444455 DOI: 10.1007/s00381-009-0884-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2009] [Indexed: 10/20/2022]
Abstract
BACKGROUND Medulloblastoma (MB) is the most common pediatric brain tumor. It is however rare in adults. The genetic and protein expression profile of medulloblastoma is complex, which is worthwhile in terms of prognostication and development or selection of targeted therapy. AIMS AND OBJECTIVES The aims and objectives to correlate the MIB-1 proliferation index and protein expression profiles of c-Myc, ERBB2, and anti-apoptotic proteins (Bcl2 and Bcl-xL) in tumor cells with histological subtypes and clinical outcome. METHODS AND MATERIAL In 50 cases, histopathological subtyping was done, and protein expression profiling by immunohistochemical technique was performed by stains for MIB-1, Bcl2, Bcl-xL, c-Myc, and ERBB2 in 30 cases. The findings were correlated with histological types and patient's average follow-up data. RESULTS Histological subtypes were similar to that described in literatures. The average expression of Bcl2, Bcl-xL, MIB-1, c-Myc, and ERBB2 were as follows: 50.38%, 38.18%, 59.03%, 46.16%, and 59.62%, respectively. Bcl2 expression showed statistically significant correlation with progress-free survival (PFS) [p = 0.046], while ERBB2 and MIB-1 showed a trend of higher expression in progressive disease. The protein expression pattern did not correlate with histological subtypes. CONCLUSION Though Bcl-2, ERBB2, and MIB-1 LI came out to be potential markers of aggressive behavior, c-Myc did not correlate with PFS in MB.
Collapse
|
26
|
Behdad A, Perry A. Central nervous system primitive neuroectodermal tumors: a clinicopathologic and genetic study of 33 cases. Brain Pathol 2009; 20:441-50. [PMID: 19725831 DOI: 10.1111/j.1750-3639.2009.00314.x] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Central nervous system (CNS) primitive neuroectodermal tumors (PNETs) include supratentorial, brain stem, and spinal cord tumors with medulloblastoma-like histopathology. The prognostic impact of various pathologic and genetic features has not been thoroughly investigated. After re-diagnosis of three infantile cases as atypical teratoid/rhabdoid tumor (AT/RT), 33 remaining CNS PNETs were retrieved for clinicopathologic and fluorescence in situ hybridization studies. Anaplastic and/or large cell features were seen in 18 of 33 (55%) examples and survival was decreased in these patients (P = 0.036). MYCN or MYCC gene amplifications were noted in about half, with a trend towards decreased survival (P = 0.112). Polysomies of chromosomes 2 and 8 were each individually associated with decreased survival in children, with an even stronger association when combined (P = 0.013). Neither EWS gene rearrangements, nor AT/RT-like 22q deletions were encountered. We conclude that in CNS PNET: (i) routine application of INI1 immunohistochemistry helps rule out AT/RT, particularly in infants; (ii) MYC gene amplifications (especially MYCN) are common; (iii) involvement of CNS parenchyma by Ewing sarcoma/peripheral PNET is rare enough that EWS gene testing is not necessary unless significant dural involvement is present; and (iv) both anaplastic/large cell features and polysomies of 2 and 8 are associated with more aggressive clinical behavior.
Collapse
Affiliation(s)
- Amir Behdad
- Department of Pathology, Cedar Sinai Medical Center, Los Angeles, Calif, USA
| | | |
Collapse
|
27
|
Aguilera DG, Das CM, Sinnappah-Kang ND, Joyce C, Taylor PH, Wen S, Hasselblatt M, Paulus W, Fuller G, Wolff JE, Gopalakrishnan V. Reactivation of death receptor 4 (DR4) expression sensitizes medulloblastoma cell lines to TRAIL. J Neurooncol 2009; 93:303-18. [PMID: 19148581 DOI: 10.1007/s11060-008-9788-x] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2008] [Accepted: 12/30/2008] [Indexed: 01/21/2023]
Abstract
OBJECT Apoptosis, a key cellular response to therapeutic agents is often inactivated in tumor cells. In this study, we evaluated the expression of the tumor necrosis family of death receptors, DR4 and DR5, in medulloblastoma tumor samples and cell lines to determine if epigenetic modulation of gene expression could sensitize tumor cell lines to TRAIL-mediated apoptosis. METHODS Human medulloblastoma samples and cell lines were analyzed for DR4 and DR5 expression by quantitative PCR and immunofluorescence assays. Cell lines with downregulated expression of one or both genes were treated with the histone deacetylase inhibitor, MS-275, and the expression of DR4 and DR5 measured by quantitative PCR, Western blotting, flow cytometry and chromatin immunoprecipitation assays. Induction of apoptosis in the presence of MS-275 was evaluated by TUNEL assay and its ability to augment TRAIL-mediated cytotoxicity was determined by MTT assays, Western blotting and flow cytometry. RESULTS Compared to normal cerebellum, DR4, but not DR5 expression was consistently downregulated in medulloblastoma tumor samples and in Daoy and D283 cell lines. Interestingly, MS-275 decreased cell growth and induced apoptosis in Daoy and D283 cells. In Daoy cells, this coincided with increased histone H3 and H4 acetylation at the DR4 promoter and enhanced DR4 gene and protein expression as well as elevated Caspase-8 activity. The involvement of DR4 in the cellular response to MS-275 was further confirmed by the observation that knockdown of DR4 and FADD abrogated apoptosis. Further, addition of TRAIL to MS-275 treated cells resulted in an enhancement of apoptosis, suggesting that the upregulated death receptors were functional. CONCLUSION Our study provides an understanding of the role of DR4 in apoptosis of medulloblastoma cell lines and suggests a potential contribution of aberrant histone deacetylation to the resistance of medulloblastoma cells to therapeutic death.
Collapse
Affiliation(s)
- Dolly G Aguilera
- Department of Pediatrics, University of Texas, M.D. Anderson Cancer Center, Houston, 77030, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Accumulation of genomic aberrations during clinical progression of medulloblastoma. Acta Neuropathol 2008; 116:383-90. [PMID: 18704466 DOI: 10.1007/s00401-008-0422-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2008] [Revised: 08/06/2008] [Accepted: 08/06/2008] [Indexed: 10/21/2022]
Abstract
Medulloblastomas comprise the most frequent malignant brain tumor in childhood and one of the biggest challenges in pediatric oncology. The current concept suggests that these tumors may undergo stepwise progression as it has been shown for other brain tumors. However, conclusive evidence of molecular progression over time has not been demonstrated yet for medulloblastoma. In the present study, 28 pairs of medulloblastoma at primary diagnosis and at the time of recurrence, either occurring as local tumor regrowth or tumor dissemination, were histopathologically and molecularly analyzed. Cytogenetic analysis included interphase fluorescence in situ hybridization for five genomic loci (MYC, MYCN, 17p, 17q, 6q) that have previously been identified as prognostic markers in primary tumors. Of 16 tumors showing early recurrence (<4 years after first diagnosis), only one showed increased histological anaplasia in the secondary lesion (6%), and two acquired genomic lesions indicative for a more malignant phenotype (13%). In contrast to this, of 12 tumors with a time to recurrence of 4 years or more, nine tumors (75%) showed a more malignant phenotype either reflected by increased anaplasia alone or by both increased anaplasia and acquirement of genomic aberrations known to be associated with inferior patient outcome. These results suggest that early recurrence in medulloblastoma mainly occurs in tumors with a highly malignant genotype and phenotype per se, whereas late recurrence is often dependent on tumor evolution toward a more malignant biology. Therefore, biopsy of recurrent tumors should be performed to assess the biologic properties of the relapsed tumor, especially when targeted therapy approaches are considered.
Collapse
|
29
|
Verma S, Tavaré CJ, Gilles FH. Histologic features and prognosis in pediatric medulloblastoma. Pediatr Dev Pathol 2008; 11:337-43. [PMID: 18201118 DOI: 10.2350/07-09-0353.1] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2007] [Accepted: 01/14/2008] [Indexed: 11/20/2022]
Abstract
Because individual histologic features in childhood medulloblastoma alter survival likelihood, the recent 4th edition of the World Health Organization (WHO) Classification of Brain Tumors recognizes desmoplastic/nodular medulloblastoma, medulloblastoma with extensive nodularity, large cell medulloblastoma, and anaplastic medulloblastoma, in addition to medulloblastoma with no other distinguishing features. To identify features affecting survival likelihood, we investigated 33 histologic features in 556 childhood tumors diagnosed as medulloblastoma in the Childhood Brain Tumor Consortium (CBTC) database; all features have CBTC verified read-reread reliability and those features important in the classification of medulloblastoma and its WHO variants regardless of their measured reliability. Nineteen features had no effect on survival likelihood, and 8 features were too prevalent or too rare to measure their effect on survival. Nodules, balls, high cell density, and fine fibrillary stroma improved survival likelihood; necrosis and prominent nucleoli worsened survival likelihood. Of note, the presence of desmoplasia, currently a defining feature (along with nodules) for desmoplastic/nodular medulloblastoma, had no effect on survival likelihood. We conclude that the presence of nodularity in medulloblastoma is important to improved survival likelihood, particularly when combined with balls and fine fibrillary stroma. Given the "overlap" of desmoplastic/nodular medulloblastoma and nodular medulloblastoma, we suggest they be combined into a diagnosis of nodular medulloblastoma, with nodules, balls, and fine fibrillary stroma as defining criteria. We also suggest that because of the considerable overlap of anaplastic medulloblastoma and large cell medulloblastoma they be combined into 1 diagnosis of anaplastic/large cell medulloblastoma, with necrosis and prominent nucleoli among the defining criteria.
Collapse
Affiliation(s)
- Shalini Verma
- Department of Pathology, LAC+USC Medical Center, Veterans Affairs Greater Los Angeles Medical Center, 1200 North State Street, Los Angeles, CA, USA
| | | | | |
Collapse
|
30
|
Abstract
The World Health Organization recently updated its classification of central nervous system tumors, adding 8 entities, as well as defining new variants and morphologic patterns of existing entities. Despite the continued refinement of brain tumor histologic classification and grading, there remain some diagnostic "gray zones" that challenge general surgical pathologists and neuropathologists alike. These include the presence of oligodendroglial features in (mixed) oligoastrocytomas and glioblastomas (GBMs), GBM variants (such as small cell GBM), meningioma classification and grading, medulloblastoma variants, ependymoma grading, the presence of "neuronal features" in otherwise morphologically classic gliomas, and low-grade gliomas with high Ki-67 labeling indices. In the current review, we discuss these issues and offer some practical guidelines for dealing with problematic cases.
Collapse
|
31
|
Entz-Werle N, Carli ED, Ducassou S, Legrain M, Grill J, Dufour C. Medulloblastoma: what is the role of molecular genetics? Expert Rev Anticancer Ther 2008; 8:1169-81. [PMID: 18588461 DOI: 10.1586/14737140.8.7.1169] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Among pediatric malignancies, medulloblastoma (MB) is one of the most common malignant tumors of the CNS. In the past few years, thanks to a multidisciplinary approach including surgery, chemo- and radiation therapy, survival has significantly improved. Despite that, a third of patients still have a low chance of being cured and long-term survivors experience severe treatment-related sequelae. MBs are usually classified according to a clinical risk stratification, based on histological features, age at diagnosis, extent of tumor resection and presence or absence of metastases. However, these clinical variables have recently been reported to be poor for defining risk-related disease. Retrospective studies have identified histological or biological factors that have distinct roles in prognosis. As several pathways have been discovered to be involved in MB pathogenesis, they should be taken into account to more accurately stratify patients and their treatment and to develop innovative therapies.
Collapse
Affiliation(s)
- Natacha Entz-Werle
- Service de Pédiatrie, U 682 Inserm CHRU Hautepierre, Avenue Molière - 67098 Strasbourg Cedex France.
| | | | | | | | | | | |
Collapse
|
32
|
Oba-Shinjo SM, Caballero OL, Jungbluth AA, Rosemberg S, Old LJ, Simpson AJG, Marie SKN. Cancer-testis (CT) antigen expression in medulloblastoma. CANCER IMMUNITY 2008; 8:7. [PMID: 18426187 PMCID: PMC2935780] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 08/16/2007] [Revised: 02/06/2008] [Accepted: 03/07/2008] [Indexed: 05/26/2023]
Abstract
Medulloblastoma is the most common childhood malignant tumor of the central nervous system. Treatment of medulloblastoma requires harmful therapy and nevertheless carries a poor prognosis. Due to their presence in various cancers and their limited expression in normal tissues, CT antigens are ideal vaccine targets for tumor immunotherapy. CT antigens, such as MAGE and NY-ESO-1, have been employed in clinical trials in various malignancies but little is known about their presence in medulloblastoma. We analyzed 25 medulloblastomas for the expression of a panel of CT antigens by RT-PCR and immunohistochemistry. Messenger RNA expression in the samples was as follows: GAGE 64%, MAGEA3/6 56%, SYCP1 44%, SLCO6A1 32%, MAGEC1 28%, MAGEC2 28%, MAGEA4 28%, NY-ESO-1 20%, MAGEA1 16%, and TPTE 0%. All cases except one (96%) were positive for mRNA expression of at least one CT gene. However, CT antigen expression was scarce on a protein level. Immunoreaction to monoclonal antibody E978 (NY-ESO-1) was negative in all cases; MA454 (MAGEA1), 57B (MAGEA4), M3H67 (MAGEA3/6), CT10#5 (MAGEC2) and #23 (GAGE) were each positive in 1 case, while the highest incidence of positive immunostaining, albeit heterogeneous, was seen with CT7-33 (MAGEC1) in 3 out of the 25 cases. The absence of correlation between mRNA and protein expression in medulloblastoma has not been observed in other tumors and further studies addressing the biology of CT antigens are necessary to investigate the present discrepant results.
Collapse
Affiliation(s)
- Sueli M Oba-Shinjo
- Department of Neurology, School of Medicine, University of São Paulo, Av Dr Arnaldo, 455, room 4110, São Paulo, Brazil
| | | | | | | | | | | | | |
Collapse
|
33
|
Newton HB, Ray-Chaudhury A, Malkin MG. Overview of Pathology and Treatment of Primary Brain Tumors. HANDBOOK OF NEURO-ONCOLOGY NEUROIMAGING 2008:9-19. [DOI: 10.1016/b978-012370863-2.50004-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/19/2023]
|
34
|
Zitterbart K, Zavrelova I, Kadlecova J, Spesna R, Kratochvilova A, Pavelka Z, Sterba J. p73 expression in medulloblastoma: TAp73/DeltaNp73 transcript detection and possible association of p73alpha/DeltaNp73 immunoreactivity with survival. Acta Neuropathol 2007; 114:641-50. [PMID: 17912537 DOI: 10.1007/s00401-007-0298-2] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2007] [Revised: 09/13/2007] [Accepted: 09/15/2007] [Indexed: 10/22/2022]
Abstract
The human p73 protein is essential for normal morphogenesis and maintenance of neural tissue. Recently, several TP73 transcripts have been revealed in medulloblastoma (MB), the most common malignant brain tumor in children. Here, we performed immunohistochemical analysis on 29 MB specimens using anti-p73alpha and anti-DeltaNp73 antibodies. Real-time PCR quantification was performed to assess TAp73 and DeltaNp73 transcripts in a subset of 13 MB samples. Normal cerebellar tissues and RNA were used for comparison. Pilot clinical-pathological correlations were also provided. We report significant differences for TAp73 and DeltaNp73 mRNA expression between tumor tissues and reference (P = 0.013, P = 0.028). Immunohistochemically, 52 and 29% MB samples were positive for p73alpha and DeltaNp73, respectively. p73alpha expression was found to be in both the nucleus and cytoplasm, whereas DeltaNp73 was localized predominantly in the cytoplasm. In normal cerebellum, positive staining for p73alpha and DeltaNp73 was observed in the Purkinje cells of newborns, not adult samples, which supports the developmental role of TP73 during organogenesis of the human cerebellum. Survival analysis has shown negative relationship of DeltaNp73-immunoreactivity with overall survival (OS) and event free survival (EFS) (P = 0.026 and P = 0.127, respectively). For p73alpha-positive cases, the negative trend in OS (P = 0.149) and EFS (P = 0.216) was also apparent. Our results indicate the involvement of p73 protein in MB tumorigenesis and define TP73 as a potential prognostic and therapeutic target for medulloblastoma.
Collapse
Affiliation(s)
- Karel Zitterbart
- Department of Pediatric Oncology, University Hospital Brno and Masaryk University, Cernopolni 9, 613 00, Brno, Czech Republic.
| | | | | | | | | | | | | |
Collapse
|
35
|
Castriconi R, Dondero A, Negri F, Bellora F, Nozza P, Carnemolla B, Raso A, Moretta L, Moretta A, Bottino C. Both CD133+ and CD133– medulloblastoma cell lines express ligands for triggering NK receptors and are susceptible to NK-mediated cytotoxicity. Eur J Immunol 2007; 37:3190-6. [DOI: 10.1002/eji.200737546] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
|
36
|
Rutkowski S, von Bueren A, von Hoff K, Hartmann W, Shalaby T, Deinlein F, Warmuth-Metz M, Soerensen N, Emser A, Bode U, Mittler U, Urban C, Benesch M, Kortmann RD, Schlegel PG, Kuehl J, Pietsch T, Grotzer M. Prognostic relevance of clinical and biological risk factors in childhood medulloblastoma: results of patients treated in the prospective multicenter trial HIT'91. Clin Cancer Res 2007; 13:2651-7. [PMID: 17473196 DOI: 10.1158/1078-0432.ccr-06-1779] [Citation(s) in RCA: 81] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE To identify better risk stratification systems in childhood medulloblastoma based on clinical factors and analysis of routinely processed formalin-fixed tumor material. EXPERIMENTAL DESIGN Formalin-fixed paraffin-embedded tumor samples from well-documented patients treated within the prospective randomized multicenter trial HIT'91 were analyzed for DNA amplification of c-myc and N-myc (n=133) and mRNA expression of c-myc and trkC (n=104; compared with human cerebellum) using validated methods of quantitative PCR and reverse transcription-PCR. Results were related to clinical data and outcome. RESULTS TrkC and c-myc mRNA expression were identified as independent prognostic factors by multivariate analysis. Three risk groups were identified. (a) Favorable risk group: all 8 patients (2 metastatic) with high trkC (>1x human cerebellum) and low c-myc mRNA expression (<or=1x human cerebellum) remained relapse-free [7-year event-free survival (EFS), 100%]. (b) Poor risk group: 10 of 15 patients with metastatic disease and high c-myc and low trkC mRNA expression relapsed (7-year EFS, 33%). (c) Intermediate risk group: the 7-year EFS of the remaining 78 patients was 65%. Among 47 M(0) stage patients, all 10 patients with high trkC mRNA expression remained relapse-free compared with 15 events in 37 patients with low trkC mRNA expression levels (7-year EFS, 100% versus 62%; P=0.056). CONCLUSIONS Whereas the collection of fresh-frozen tumor samples remains a major challenge in large clinical trials, routinely processed paraffin-embedded tissue samples can be used to quantitate the prognostic biological markers trkC and c-myc. On prospective validation of cutoff levels, this may lead to improved stratification of treatment for children with medulloblastoma.
Collapse
Affiliation(s)
- Stefan Rutkowski
- University Children's Hospital of Wuerzburg, Wuerzburg, Germany.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Polkinghorn WR, Tarbell NJ. Medulloblastoma: tumorigenesis, current clinical paradigm, and efforts to improve risk stratification. ACTA ACUST UNITED AC 2007; 4:295-304. [PMID: 17464337 DOI: 10.1038/ncponc0794] [Citation(s) in RCA: 140] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2006] [Accepted: 12/22/2006] [Indexed: 11/08/2022]
Abstract
Medulloblastoma is the most common brain malignancy in children and tremendous advances have recently been made in understanding the pathogenesis of this tumor. The Hedgehog and Wingless signaling pathways are implicated in medulloblastoma development, and both pathways were discovered as a result of analyses of genetic syndromes associated with the tumor. Over the past 80 years, considerable progress has been made in the treatment of what was once a fatal disease. The first survival reports followed the introduction of craniospinal irradiation, and yet the success of this modality, which continues to be a central component of treatment regimens for patients older than 3 years, comes at a significant cost. The present challenge in medulloblastoma treatment is to improve upon existing survival rates and to minimize the side effects of treatment. The current tools of clinical risk assessment fail to adequately identify patients older than 3 years who require less radiation and those who require more radiation. Significant effort has been made to improve clinical risk stratification and titration of treatment by analyzing properties of the tumor cells themselves for prognostic significance. These efforts include identifying histopathologic, cytogenetic, and molecular features that may correlate with prognosis.
Collapse
|
38
|
Yang CS, Jan YJ, Wang J, Shen CC, Chen CCC, Chen M. Spinal atypical teratoid/rhabdoid tumor in a 7-year-old boy. Neuropathology 2007; 27:139-44. [PMID: 17494515 DOI: 10.1111/j.1440-1789.2007.00752.x] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Reported herein is an unusual case of atypical teratoid/ rhabdoid tumor (AT/RT) of the lumbar spine with an intradural extramedullary location in a 7-year-old boy. Histologically, this tumor contained rhabdoid cells, pale cells, and sickle-shaped embracing cells without primitive neuroectodermal tumor (PNET), mesenchymal or epithelial components. Immunohistochemical staining showed that these tumor cells react positively for epithelial membrane antigen (EMA), vimentin, cytokeratin (AE1/AE3), CD99 and neurofilament protein, but negatively for INI1 antibody. Chromosome 22q deletion was demonstrated on fluorescence in situ hybridization.
Collapse
Affiliation(s)
- Chii-Shuenn Yang
- Department of Pathology, Taichung Veterans General Hospital, College of Medicine, Chung Shan Medical University, Taichung, Taiwan.
| | | | | | | | | | | |
Collapse
|
39
|
Verlooy J, Mosseri V, Bracard S, Tubiana AL, Kalifa C, Pichon F, Frappaz D, Chastagner P, Pagnier A, Bertozzi AI, Gentet JC, Sariban E, Rialland X, Edan C, Bours D, Zerah M, Le Gales C, Alapetite C, Doz F. Treatment of high risk medulloblastomas in children above the age of 3 years: A SFOP study. Eur J Cancer 2006; 42:3004-14. [PMID: 16956759 DOI: 10.1016/j.ejca.2006.02.026] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2005] [Revised: 02/21/2006] [Accepted: 02/21/2006] [Indexed: 10/24/2022]
Abstract
AIM Improvement of EFS of children older than 3 years with high risk medulloblastoma. METHODS Between 1993 and 1999, 115 patients (3-18 years, mean 8 years) with high risk medulloblastoma were included. After surgery treatment consisted of chemotherapy ('8in1' and etoposide/carboplatin) before and after craniospinal radiotherapy. RESULTS Patients were staged using Chang-criteria (PF residue only, M1 and M2/M3) by local investigator as well as by central review panel (82.4% concordance). Chemotherapy was well tolerated without major delays in radiotherapy. With a mean follow up of 81 months (9-119), 5-year EFS was 49.8% and OS 60.1%. In detail according to subgroups EFS was 68.8% for PF residue only, 58.8% for M1 disease and 43.1% for M2/M3. CONCLUSION M1 patients are legitimate high risk patients. Survival rates are still very low for high risk medulloblastoma patients and future trials should therefore focus on more intensive (chemotherapy/radiotherapy) treatment.
Collapse
Affiliation(s)
- J Verlooy
- Department of Paediatric Oncology, Institut Curie, Service d'Oncologie Pediatrique, 26 rue d'Ulm, 75231 Paris Cedex 05, France
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Shakhova O, Leung C, van Montfort E, Berns A, Marino S. Lack of Rb and p53 delays cerebellar development and predisposes to large cell anaplastic medulloblastoma through amplification of N-Myc and Ptch2. Cancer Res 2006; 66:5190-200. [PMID: 16707443 DOI: 10.1158/0008-5472.can-05-3545] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Medulloblastomas are among the most common malignant brain tumors in childhood. They typically arise from neoplastic transformation of granule cell precursors in the cerebellum via deregulation of molecular pathways involved in normal cerebellar development. In a mouse model, we show here that impairment of the balance between proliferation and differentiation of granule cell precursors in the external granular layer of the developing cerebellum predisposes but is not sufficient to induce neoplastic transformation of these progenitor cells. Using array-based chromosomal comparative genomic hybridization, we show that genetic instability resulting from inactivation of the p53 pathway together with deregulation of proliferation induced by Rb loss eventually leads to neoplastic transformation of these cells by acquiring additional genetic mutations, mainly affecting N-Myc and Ptch2 genes. Moreover, we show that p53 loss influences molecular mechanisms that cannot be mimicked by the loss of either p19(ARF), p21, or ATM.
Collapse
Affiliation(s)
- Olga Shakhova
- Institute of Clinical Pathology, University Hospital, Zurich, Switzerland
| | | | | | | | | |
Collapse
|
41
|
Kochbati L, Bouaouina N, Hentati D, Nasr C, Besbes M, Benna F, Boussen H, Maalej M. [Medulloblastoma with extracentral nervous system metastases: clinical presentation and risk factors]. Cancer Radiother 2006; 10:107-11. [PMID: 16600659 DOI: 10.1016/j.canrad.2006.02.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2005] [Revised: 02/13/2006] [Accepted: 02/28/2006] [Indexed: 10/24/2022]
Abstract
PURPOSE Extra-central nervous system (extra-CNS) metastases are relatively unknown failure patterns in medulloblastoma. The aim of this study was to analyse epidemiological, clinical and aetiopathological aspects of these extra-CNS localisations. PATIENTS AND METHODS Extra-CNS metastases were retrospectively identified in patients treated in the department of radiation therapy at Salah-Azaïz institute (ISA) for medulloblastoma. These metastases were diagnosed as extra-CNS for all secondary localisations not related to other tumour aetiology. Aetiopathological aspects are discussed with a literature review. RESULTS Among 103 patients treated and followed-up in the department of radiation therapy of ISA from 1970 to 1992, 8 developed extra-CNS metastases (7.7%). Age at diagnosis of primitive tumour varied from 3 to 23 years. Sex ratio was 1. Primitive tumour treatment was: complete surgical resection in 4 patients with preoperative cerebrospinal fluid shunting in two, cerebrospinal axis irradiation in 7 patients and a cerebral-limited irradiation in 1. Two patients received chemotherapy for their initial treatment (systemic in one case and intrathecal in the other). The mean free-interval from diagnosis of primitive tumour to extra-CNS metastases was 23 months, varying from 8 to 53 months. These metastases were located in the liver (1 case), cervical lymph nodes (2 cases), bone marrow (1 case) and bone (2 cases). Two patients had multiple metastases: bone and bone marrow (in one), lung, pleura, cervical lymph node and bone localisations (in one). Treatment of these metastases was: chemotherapy in 5 cases, chemotherapy and radiation in one, radiation therapy in one and 2 patients were given only supportive care treatment. All patients died or are in progressive disease in less than one year from the diagnosis of extra-CNS metastases. CONCLUSION Extra-CNS metastases are not rare and have a poor prognosis. The most commonly involved sites are bone, cervical lymph nodes and bone marrow. A complete work-up at initial diagnosis is recommended to screen early metastases. Literature review showed that histopathologic grading might help to identify groups at risk.
Collapse
Affiliation(s)
- Lotfi Kochbati
- Radiotherapy department, Salah-Azaïz institute, boulevard Bab-Saadoun, Tunis, Tunisia. lotfi@
| | | | | | | | | | | | | | | |
Collapse
|
42
|
Min HS, Lee YJ, Park K, Cho BK, Park SH. Medulloblastoma: histopathologic and molecular markers of anaplasia and biologic behavior. Acta Neuropathol 2006; 112:13-20. [PMID: 16691420 DOI: 10.1007/s00401-006-0073-9] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2005] [Revised: 04/03/2006] [Accepted: 04/04/2006] [Indexed: 12/31/2022]
Abstract
Large cell/anaplastic (LC/A) medulloblastoma (MB) is a recently recognized variant of medulloblastoma known to be associated with an advanced stage and a poor prognosis. Although Eberhart et al. suggested histopathologic grading of medulloblastoma in 2002, no consensus has been reached in terms of determining the criteria of an LC/A variant, and its biological behavior continues to be the subject of debate. We retrospectively analyzed 74 cases (range 0.25-15 years) of MB clinicopathologically using the criteria established by Eberhart et al. The LC/A variant was identified in 16 cases (22% of MB cases), five of which showed a poor outcome. Most LC/A variant cases revealed synaptophysin immunoexpression (75%), but no epidermal growth factor receptor (EGFR) expression. Expression of synaptophysin, NeuN, GFAP, p53, c-erbB2, and EGFR did not differ in LC/A and non-LC/A variants. Seven of the 74 cases of medulloblastoma showed erbB2 amplification by FISH, four of which were LC/A variants. N-myc amplification was observed in only one LC/A variant, but no c-myc amplification was found. In patients younger than 10 years, the LC/A variant showed a significantly poorer outcome than the non-LC/A variant (P = 0.02), while no difference was found in older patients. Multivariate analysis revealed only metastasis on MRI and p53 expression, but not anaplasia as unfavorable prognostic factors. Our study suggests that prognostic implications of anaplasia in medulloblastoma are uncertain, and that the reproducibility of the histopathologic criteria of the LC/A variant should be reassessed before they can be applied in practical use.
Collapse
Affiliation(s)
- Hye Sook Min
- Department of Pathology, Seoul National University College of Medicine, 28 Yongon-dong, Chongno-gu, Seoul, 110-799, Korea
| | | | | | | | | |
Collapse
|
43
|
Su X, Gopalakrishnan V, Stearns D, Aldape K, Lang FF, Fuller G, Snyder E, Eberhart CG, Majumder S. Abnormal expression of REST/NRSF and Myc in neural stem/progenitor cells causes cerebellar tumors by blocking neuronal differentiation. Mol Cell Biol 2006; 26:1666-78. [PMID: 16478988 PMCID: PMC1430235 DOI: 10.1128/mcb.26.5.1666-1678.2006] [Citation(s) in RCA: 119] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Medulloblastoma, one of the most malignant brain tumors in children, is thought to arise from undifferentiated neural stem/progenitor cells (NSCs) present in the external granule layer of the cerebellum. However, the mechanism of tumorigenesis remains unknown for the majority of medulloblastomas. In this study, we found that many human medulloblastomas express significantly elevated levels of both myc oncogenes, regulators of neural progenitor proliferation, and REST/NRSF, a transcriptional repressor of neuronal differentiation genes. Previous studies have shown that neither c-Myc nor REST/NRSF alone could cause tumor formation. To determine whether c-Myc and REST/NRSF act together to cause medulloblastomas, we used a previously established cell line derived from external granule layer stem cells transduced with activated c-myc (NSC-M). These immortalized NSCs were able to differentiate into neurons in vitro. In contrast, when the cells were engineered to express a doxycycline-regulated REST/NRSF transgene (NSC-M-R), they no longer underwent terminal neuronal differentiation in vitro. When injected into intracranial locations in mice, the NSC-M cells did not form tumors either in the cerebellum or in the cerebral cortex. In contrast, the NSC-M-R cells did produce tumors in the cerebellum, the site of human medulloblastoma formation, but not when injected into the cerebral cortex. Furthermore, the NSC-M-R tumors were blocked from terminal neuronal differentiation. In addition, countering REST/NRSF function blocked the tumorigenic potential of NSC-M-R cells. To our knowledge, this is the first study in which abnormal expression of a sequence-specific DNA-binding transcriptional repressor has been shown to contribute directly to brain tumor formation. Our findings indicate that abnormal expression of REST/NRSF and Myc in NSCs causes cerebellum-specific tumors by blocking neuronal differentiation and thus maintaining the "stemness" of these cells. Furthermore, these results suggest that such a mechanism plays a role in the formation of human medulloblastoma.
Collapse
Affiliation(s)
- Xiaohua Su
- Department of Molecular Genetics, University of Texas M. D. Anderson Cancer Center, 1515 Holcombe Boulevard, Mail 1006, Room S13.8136C, Houston, TX 77030, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Stearns D, Chaudhry A, Abel TW, Burger PC, Dang CV, Eberhart CG. c-myc overexpression causes anaplasia in medulloblastoma. Cancer Res 2006; 66:673-81. [PMID: 16423996 DOI: 10.1158/0008-5472.can-05-1580] [Citation(s) in RCA: 87] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Both anaplasia and increased c-myc gene expression have been shown to be negative prognostic indicators for survival in medulloblastoma patients. myc gene amplification has been identified in many large cell/anaplastic medulloblastoma, but no causative link between c-myc and anaplastic changes has been established. To address this, we stably overexpressed c-myc in two medulloblastoma cell lines, DAOY and UW228, and examined the changes in growth characteristics. When analyzed in vitro, cell lines with increased levels of c-myc had higher rates of growth and apoptosis as well as significantly improved ability to form colonies in soft agar compared with control. When injected s.c. into nu/nu mice, flank xenograft tumors with high levels of c-myc in DAOY cell line background were 75% larger than those derived from control. Overexpression of c-myc was required for tumor formation by UW228 cells. Most remarkably, the histopathology of the Myc tumors was severely anaplastic, with large areas of necrosis/apoptosis, increased nuclear size, and macronucleoli. Indices of proliferation and apoptosis were also significantly higher in Myc xenografts. Thus, c-myc seems to play a causal role in inducing anaplasia in medulloblastoma. Because anaplastic changes are often observed in recurrent medulloblastoma, we propose that c-myc dysregulation is involved in the progression of these malignant embryonal neoplasms.
Collapse
Affiliation(s)
- Duncan Stearns
- Department of Neuropathology, Johns Hopkins University School of Medicine, 558 Ross Building, 720 Rutland Avenue, Baltimore, MD 21205, USA.
| | | | | | | | | | | |
Collapse
|
45
|
Bobola MS, Finn LS, Ellenbogen RG, Geyer JR, Berger MS, Braga JM, Meade EH, Gross ME, Silber JR. Apurinic/apyrimidinic endonuclease activity is associated with response to radiation and chemotherapy in medulloblastoma and primitive neuroectodermal tumors. Clin Cancer Res 2006; 11:7405-14. [PMID: 16243814 DOI: 10.1158/1078-0432.ccr-05-1068] [Citation(s) in RCA: 77] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Apurinic/apyrimidinic endonuclease (Ap endo) is a key DNA repair activity that confers resistance to radiation- and alkylator-induced cytotoxic abasic sites in human cells. We assayed apurinic/apyrimidinic endonuclease activity in medulloblastomas and primitive neuroectodermal tumors (PNET) to establish correlates with tumor and patient characteristics and with response to adjuvant radiation plus multiagent chemotherapy. EXPERIMENTAL DESIGN Ap endo activity was assayed in 52 medulloblastomas and 10 PNETs from patients 0.4 to 21 years old. Ape1/Ref-1, the predominant human Ap endo activity, was measured in 42 medulloblastomas by immunostaining. Cox proportional hazards regression models were used to analyze the association of activity with time to tumor progression (TTP). RESULTS Tumor Ap endo activity varied 180-fold and was significantly associated with age and gender. Tumor Ape1/Ref-1 was detected almost exclusively in nuclei. In a multivariate model, with Ap endo activity entered as a continuous variable, the hazard ratio for progression after adjuvant treatment in 46 medulloblastomas and four PNETs increased by a factor of 1.073 for every 0.01 unit increase in activity (P < or = 0.001) and was independent of age and gender. Suppressing Ap endo activity in a human medulloblastoma cell line significantly increased sensitivity to 1,3-bis(2-chlororethyl)-1-nitrosourea and temozolomide, suggesting that the association of tumor activity with TTP reflected, at least in part, abasic site repair. CONCLUSIONS Our data (a) suggest that Ap endo activity promotes resistance to radiation plus chemotherapy in medulloblastomas/PNETs, (b) provide a potential marker of treatment outcome, and (c) suggest clinical use of Ap endo inhibitors to overcome resistance.
Collapse
MESH Headings
- Adolescent
- Adult
- Antineoplastic Agents, Alkylating/pharmacology
- Blotting, Western
- Brain/drug effects
- Brain/enzymology
- Brain/radiation effects
- Brain Neoplasms/enzymology
- Brain Neoplasms/pathology
- Brain Neoplasms/therapy
- Carmustine/pharmacology
- Cell Line, Tumor
- Cell Nucleus/enzymology
- Cell Survival/drug effects
- Cell Survival/genetics
- Child
- Child, Preschool
- Combined Modality Therapy
- DNA-(Apurinic or Apyrimidinic Site) Lyase/genetics
- DNA-(Apurinic or Apyrimidinic Site) Lyase/metabolism
- Disease Progression
- Dose-Response Relationship, Drug
- Female
- Humans
- Immunohistochemistry
- Infant
- Infant, Newborn
- Male
- Medulloblastoma/enzymology
- Medulloblastoma/pathology
- Medulloblastoma/therapy
- Multivariate Analysis
- Neuroectodermal Tumors, Primitive/enzymology
- Neuroectodermal Tumors, Primitive/pathology
- Neuroectodermal Tumors, Primitive/therapy
- Oligonucleotides, Antisense/genetics
- RNA, Small Interfering/genetics
- Time Factors
- Transfection
Collapse
Affiliation(s)
- Michael S Bobola
- Division of Neurosurgery, Department of Surgery, Children's Hospital and Regional Medical Center, Seattle, WA 98105, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Newton HB, Ray-Chaudhury A. Overview of Brain Tumor Epidemiology and Histopathology. HANDBOOK OF BRAIN TUMOR CHEMOTHERAPY 2006:3-20. [DOI: 10.1016/b978-012088410-0/50039-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/19/2023]
|
47
|
Ellison DW, Onilude OE, Lindsey JC, Lusher ME, Weston CL, Taylor RE, Pearson AD, Clifford SC. β-Catenin Status Predicts a Favorable Outcome in Childhood Medulloblastoma: The United Kingdom Children's Cancer Study Group Brain Tumour Committee. J Clin Oncol 2005; 23:7951-7. [PMID: 16258095 DOI: 10.1200/jco.2005.01.5479] [Citation(s) in RCA: 305] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Purpose Identifying pathobiological correlates of clinical behavior or therapeutic response currently represents a key challenge for medulloblastoma research. Nuclear accumulation of the β-catenin protein is associated with activation of the Wnt/Wg signaling pathway, and mutations affecting components of this pathway have been reported in approximately 15% of sporadic medulloblastomas. We tested the hypothesis that nuclear immunoreactivity for β-catenin is a prognostic marker in medulloblastoma, and assessed the relationship between nuclear β-catenin immunoreactivity and mutations of CTNNB1 and APC. Patients and Methods Medulloblastomas from children entered onto the International Society for Pediatric Oncology (SIOP)/United Kingdom Children's Cancer Study Group (UKCCSG) PNET3 trial (n = 109) were examined for β-catenin immunoreactivity, and where tissue was available, evidence of CTNNB1 and APC mutations. The results were correlated with clinicopathologic variables, principally outcome. Results Children with medulloblastomas that showed a nucleopositive β-catenin immunophenotype (27 of 109; 25%) had significantly better overall (OS) and event-free (EFS) survivals than children with tumors that showed either membranous/cytoplasmic β-catenin immunoreactivity or no immunoreactivity (P = .0015 and P = .0026, respectively). For β-catenin nucleopositive and nucleonegative medulloblastomas, 5-year OS was 92.3% (95% CI, 82% to 100%) versus 65.3% (95% CI, 54.8 to 75.7%), and 5-year EFS was 88.9% (95% CI, 77% to 100%) versus 59.5% (95% CI, 48.8 to 70.2%), respectively. Mutations in CTNNB1 were found exclusively among medulloblastomas that demonstrated nuclear β-catenin immunoreactivity, but no evidence of APC mutation was found in these cases. All children with β-catenin nucleopositive large cell/anaplastic medulloblastomas and β-catenin nucleopositive medulloblastomas presenting with metastatic disease are alive at least 5 years postdiagnosis. Conclusion Nuclear accumulation of β-catenin appears to be a marker of favorable outcome in medulloblastoma, and should be investigated further in large group-wide trials.
Collapse
Affiliation(s)
- David W Ellison
- Northern Institute for Cancer Research, University of Newcastle, UK.
| | | | | | | | | | | | | | | |
Collapse
|
48
|
Misaki K, Marukawa K, Hayashi Y, Fukusato T, Minamoto T, Hasegawa M, Yamashita J, Fujisawa H. Correlation of gamma-catenin expression with good prognosis in medulloblastomas. J Neurosurg 2005; 102:197-206. [PMID: 16156230 DOI: 10.3171/jns.2005.102.2.0197] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
OBJECT Medulloblastoma is a malignant cerebellar tumor of childhood and is difficult to cure due to frequent cerebrospinal fluid dissemination. Amplification of the c-myc gene (4%) and messenger (mRNA) overexpression (50%) are known to be adverse prognostic indicators. Because mRNA overexpression cannot be explained by gene amplification alone, mechanisms other than gene amplification are postulated. Molecules on the Wnt signal pathway in primary tumors were examined. METHODS Immunohistochemical and cytogenetic examinations of beta- and gamma-catenin, c-myc, N-myc, and cyclin D1 in 24 primary medulloblastomas were conducted, and their clinical relevance was evaluated. Cytoplasmic/membranous staining of beta- and gamma-catenin was detected in 19 (79%) and nine (37%) cases, respectively, and nuclear expression of cyclin D1 and c-myc was detected in six (25%) and 21 (83%) cases, respectively. The expression levels of gamma-catenin in Western blot analysis and immunohistochemistry were similar. By differential polymerase chain reaction, c-myc and N-myc were amplified separately in two large cell/anaplastic medulloblastomas. No cyclin D1 amplification, or beta- or gamma-catenin mutations were found. Kaplan-Meier analysis revealed no dissemination at diagnosis (Chang Grade M0) and gamma-catenin expression was correlated with good prognosis (p = 0.0002 and 0.003, respectively). Expression of gamma-catenin was also significant in the M0 group (p = 0.022). Expression of cyclin D1 showed a trend toward adverse outcome (p = 0.057) and all patients in whom cyclin D1 expression was found died of disease. CONCLUSIONS Expression of gamma-catenin is of great prognostic value and its immunohistochemistry may be useful for further stratification of treatment. Cyclin D expression may have the potential to be an adverse prognostic indicator.
Collapse
Affiliation(s)
- Koichi Misaki
- Department of Neurosurgery, Graduate School of Medical Science, Division of Diagnostic Molecular Oncology, Cancer Research Institute, Kanazawa University, Kanazawa, Japan
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Huang A, Ho CSW, Ponzielli R, Barsyte-Lovejoy D, Bouffet E, Picard D, Hawkins CE, Penn LZ. Identification of a novel c-Myc protein interactor, JPO2, with transforming activity in medulloblastoma cells. Cancer Res 2005; 65:5607-19. [PMID: 15994933 DOI: 10.1158/0008-5472.can-05-0500] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
c-myc oncogene activation is critical in the pathogenesis of a spectrum of human malignancies. The c-Myc NH2-terminal domain (MycNTD) is essential for cellular transformation, and mediates critical protein interactions that modulate c-Myc oncogenic properties. In medulloblastoma, the most common malignant pediatric brain tumor, deregulated c-myc expression is linked with poorer disease phenotypes and outcomes. The biological basis for these associations is, however, not well understood. To better understand mechanisms underlying Myc-mediated transformation of medulloblastoma, we sought to identify novel MycNTD protein interactors from a medulloblastoma cell line library using a unique two-hybrid system. We identified a novel MycNTD binding protein, JPO2, which shows nuclear colocalization with c-Myc, and interacts with c-Myc both in vitro and in mammalian cells. In Rat1a transformation assays, JPO2 potentiates c-Myc transforming activity, and can complement a transformation-defective Myc mutant. Immunohistochemical studies indicate tumor-specific JPO2 expression in human medulloblastoma, and an association of JPO2 expression with metastatic tumors. Significantly, JPO2 expression induces colony formation in UW228, a medulloblastoma cell line, whereas RNAi-mediated JPO2 knockdown impairs colony formation in UW228, and in Myc-transformed UW228 cells. These data provide evidence for biochemical and functional interaction between c-Myc and JPO2 in medulloblastoma transformation. JPO2 is closely related to JPO1, a Myc transcriptional target with transforming activity. As tumor-specific JPO1 expression in human and murine medulloblastoma has also been reported; these collective observations suggest important functional links between the novel JPO protein family and c-Myc in medulloblastoma transformation.
Collapse
|
50
|
Oyharcabal-Bourden V, Kalifa C, Gentet JC, Frappaz D, Edan C, Chastagner P, Sariban E, Pagnier A, Babin A, Pichon F, Neuenschwander S, Vinchon M, Bours D, Mosseri V, Le Gales C, Ruchoux M, Carrie C, Doz F. Standard-Risk Medulloblastoma Treated by Adjuvant Chemotherapy Followed by Reduced-Dose Craniospinal Radiation Therapy: A French Society of Pediatric Oncology Study. J Clin Oncol 2005; 23:4726-34. [PMID: 16034048 DOI: 10.1200/jco.2005.00.760] [Citation(s) in RCA: 101] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Objective The primary objective of this study was to decrease the late effects of prophylactic radiation without reducing survival in standard-risk childhood medulloblastoma. Patients and Methods Inclusion criteria were as follows: children between the ages of 3 and 18 years with total or subtotal tumor resection, no metastasis, and negative postoperative lumbar puncture CSF cytology. Two courses of eight drugs in 1 day followed by two courses of etoposide plus carboplatin (500 and 800 mg/m2 per course, respectively) were administered after surgery. Radiation therapy had to begin 90 days after surgery. Delivered doses were 55 Gy to the posterior fossa and 25 Gy to the brain and spinal canal. Results Between November 1991 and June 1998, 136 patients (median age, 8 years; median follow-up, 6.5 years) were included. The overall survival rate and 5-year recurrence-free survival rate were 73.8% ± 7.6% and 64.8% ± 8.1%, respectively. Radiologic review showed that 4% of patients were wrongly included. Review of radiotherapy technical files demonstrated a correlation between the presence of a major protocol deviation and treatment failure. The 5-year recurrence-free survival rate of patients included in this study with all optimal quality controls of histology, radiology, and radiotherapy was 71.8% ± 10.5%. In terms of sequelae, 31% of patients required growth hormone replacement therapy and 25% required special schooling. Conclusion Reduced-dose craniospinal radiation therapy can be proposed in standard-risk medulloblastoma provided staging and radiation therapy are performed under optimal conditions.
Collapse
|